InteractiveFly: GeneBrief

scribbled: Biological Overview | Regulation | Developmental Biology | Effects of Mutation | Evolutionary Homologs | References


Gene name - scribbled

Synonyms - scribble

Cytological map position - 97B8-9

Function - scaffold protein

Keywords - apical/basal polarity, epidermis

Symbol - scrib

FlyBase ID: FBgn0263289

Genetic map position -

Classification - Multiple PDZ with LLR regions

Cellular location - cytoplasmic



NCBI link: Entrez Gene

scrib orthologs: Biolitmine

Recent literature
Felix, M., Chayengia, M., Ghosh, R., Sharma, A. and Prasad, M. (2015). dPak3 regulates apical-basal polarity in migrating border cells during Drosophila oogenesis. Development [Epub ahead of print]. PubMed ID: 26395489
Summary:
During group cell migration collectively moving cells are physically attached to each other and retain some degree of apical-basal polarity during the migratory phase. Though much is known about direction sensing, it is far from clear how polarity is regulated in diverse instances of multicellular movement. This study reports the role of dPak3, a group I p21 activated serine-threonine protein kinase, in maintaining apical-basal polarity in migrating border cell cluster during Drosophila oogenesis. dPak3 is enriched in border cells and downregulation of its function impedes border cell movement. Time-lapse imaging suggests that dPak3 affects protrusive behavior of the border cell cluster specifically regulating the stability and directionality of the protrusions. This study shows that dPak3 functions downstream of the guidance receptor signaling to regulate the level and distribution of F-actin in the migrating border cells. Further evidence is provided that dPak3 genetically interacts with lateral polarity marker Scribble and regulates c-Jun N-terminal kinase (JNK) signaling in the moving border cells. Since dPak3 depletion results in mislocalization of several apical-basal polarity markers including Stardust, Crumbs and Coracle and overexpression of D-jun rescues the polarity of the dPak3-depleted cluster, it is proposed that dPak3 functions through the JNK signaling to modulate the apical-basal polarity of the migrating border cell cluster. Interestingly, loss of apical-basal polarity was also observed in Rac1 depleted border cell cluster suggesting that the guidance receptor signaling functions through Rac GTPase and dPak3 to regulate overall polarity of the cluster to mediate efficient collective movement of the border cells to the oocyte boundary.

Zhou, Y., Chang, R., Ji, W., Wang, N., Qi, M., Xu, Y., Guo, J. and Zhan, L. (2015). Loss of Scribble promotes Snail translation through translocation of HuR and enhances cancer drug resistance. J Biol Chem [Epub ahead of print]. PubMed ID: 26527679
Summary:
Drug resistance of cancer cells to various therapeutic agents and molecular targets is a major problem facing current cancer research. The tumor suppressor gene Scribble encodes a polarity protein that is conserved between Drosophila and mammals; loss of the locus disrupts cell polarity, inhibits apoptosis, and mediates cancer process. However, the role of Scribble in drug resistance remains unknown. This study shows that knockdown of Scribble enhances drug resistance by permitting accumulation of Snail, which functions as a transcription factor during the epithelial-mesenchymal transition. Then, loss of Scribble activates the mRNA binding protein HuR (ELAV in Drosophila) by facilitating translocation of HuR from the nucleus to the cytoplasm. Furthermore, HuR can recognize AU-rich elements (AREs) of the Snail-encoding mRNA, thereby regulating Snail translation. Moreover, Loss of Scribble induced HuR translocation mediates the accumulation of Snail via activation of the p38 MAPK pathway. Thus, this work clarifies the role of polarity protein Scribble, which is directly implicated in the regulation of developmental transcription factor Snail, and suggesting a mechanism for Scribble mediating cancer drug resistance.

Cervantes-Sandoval, I., Chakraborty, M., MacMullen, C. and Davis, R.L. (2016). Scribble scaffolds a signalosome for active forgetting. Neuron 90(6):1230-1242. PubMed ID: 27263975
Summary:
Forgetting, one part of the brain's memory management system, provides balance to the encoding and consolidation of new information by removing unused or unwanted memories or by suppressing their expression. Recent studies have identified the small G protein, Rac1, as a key player in the Drosophila mushroom bodies neurons (MBn) for active forgetting. It has also been shown that a few dopaminergic neurons (DAn) that innervate the MBn mediate forgetting. This study shows that Scribble, a scaffolding protein known primarily for its role as a cell polarity determinant, orchestrates the intracellular signaling for normal forgetting. Knocking down scribble expression in either MBn or DAn impairs normal memory loss. Scribble interacts physically and genetically with Rac1, Pak3, and Cofilin within MBn, nucleating a forgetting signalosome that is downstream of dopaminergic inputs that regulate forgetting. These results bind disparate molecular players in active forgetting into a single signaling pathway: Dopamine→ Dopamine Receptor→ Scribble→ Rac→ Cofilin.

Waghmare, I. and Kango-Singh, M. (2016). Loss of cell adhesion increases tumorigenic potential of polarity deficient Scribble mutant cells. PLoS One 11: e0158081. PubMed ID: 27327956
Summary:
Epithelial polarity genes are important for maintaining tissue architecture, and regulating growth. The Drosophila neoplastic tumor suppressor gene scribble (scrib) belongs to the basolateral polarity complex. Loss of scrib results in disruption of its growth regulatory functions, and downregulation or mislocalization of Scrib is correlated to tumor growth. Somatic scribble mutant cells (scrib-) surrounded by wild-type cells undergo apoptosis, which can be prevented by introduction of secondary mutations that provide a growth advantage. Using genetic tools in Drosophila, the phenotypic effects were analyzed of loss of scrib in different growth promoting backgrounds. This study investigated if a central mechanism that regulates cell adhesion governs the growth and invasive potential of scrib mutant cells. Increased proliferation, and survival abilities of scrib- cells in different genetic backgrounds affect their differentiation, and intercellular adhesion. Further, loss of scrib is sufficient to cause reduced cell survival, activation of the JNK pathway and a mild reduction of cell adhesion. These data show that for scrib cells to induce aggressive tumor growth characterized by loss of differentiation, cell adhesion, increased proliferation and invasion, cooperative interactions that derail signaling pathways play an essential role in the mechanisms leading to tumorigenesis. Thus, this study provides new insights on the effects of loss of scrib and the modification of these effects via cooperative interactions that enhance the overall tumorigenic potential of scrib deficient cells.
Schimizzi, G. V., Maher, M. T., Loza, A. J. and Longmore, G. D. (2016). Disruption of the Cdc42/Par6/aPKC or Dlg/Scrib/Lgl polarity complex promotes epithelial proliferation via overlapping mechanisms. PLoS One 11: e0159881. PubMed ID: 27454609
Summary:
The establishment and maintenance of apical-basal polarity is a defining characteristic and essential feature of functioning epithelia. Apical-basal polarity (ABP) proteins are also tumor suppressors that are targeted for disruption by oncogenic viruses and are commonly mutated in human carcinomas. Using the proliferating Drosophila wing disc epithelium, this study demonstrates that disruption of the junctional [Cdc42/Par6/Par3/Atypical PKC (aPKC)] complex vs. basolateral polarity complex [Scribble (Scrib)/Discs Large (Dlg)/Lethal Giant Larvae (Lgl)] complex results in increased epithelial proliferation via distinct downstream signaling pathways. Disruption of the basolateral polarity complex results in JNK-dependent proliferation, while disruption of the junctional complex primarily results in p38-dependent proliferation. Surprisingly, the Rho-Rok-Myosin contractility apparatus appears to play opposite roles in the regulation of the proliferative phenotype based on which polarity complex is disrupted. In contrast, non-autonomous Tumor Necrosis Factor (TNF; Eiger) signaling appears to suppress the proliferation that results from apical-basal polarity disruption, regardless of which complex is disrupted. Finally it was demonstrated that disruption of the junctional polarity complex activates JNK via the Rho-Rok-Myosin contractility apparatus independent of the cortical actin regulator, Moesin.
Vaughen, J. and Igaki, T. (2016). Slit-Robo repulsive signaling extrudes tumorigenic cells from epithelia. Dev Cell 39: 683-695. PubMed ID: 27997825
Summary:
Cells dynamically interact throughout animal development to coordinate growth and deter disease. For example, cell-cell competition weeds out aberrant cells to enforce homeostasis. In Drosophila, tumorigenic cells mutant for the cell polarity gene scribble (scrib) are actively eliminated from epithelia when surrounded by wild-type cells. While scrib cell elimination depends critically on JNK signaling, JNK-dependent cell death cannot sufficiently explain scrib cell extirpation. Thus, how JNK executed cell elimination remained elusive. This study shows that repulsive Slit-Robo2-Ena signaling exerts an extrusive force downstream of JNK to eliminate scrib cells from epithelia by disrupting E-cadherin. While loss of Slit-Robo2-Ena in scrib cells potentiates scrib tumor formation within the epithelium, Robo2-Ena hyperactivation surprisingly triggers luminal scrib tumor growth following excess extrusion. This extrusive signaling is amplified by a positive feedback loop between Slit-Robo2-Ena and JNK. These observations provide a potential causal mechanism for Slit-Robo dysregulation in numerous human cancers.

Rives-Quinto, N., Franco, M., de Torres-Jurado, A. and Carmena, A. (2017). canoe and scribble loss synergizes causing tumor-like overgrowth via Ras activation in neural stem cells and epithelia. Development [Epub ahead of print]. PubMed ID: 28619817
Summary:
Over the past decade an intriguing connection between asymmetric cell division, stem cells and tumorigenesis has emerged. Neuroblasts, the neural stem cells of the Drosophila central nervous system, divide asymmetrically and constitute an excellent paradigm for further investigating that connection. This study shows that the simultaneous loss of the asymmetric cell division regulators Canoe (Afadin in mammals) and Scribble in neuroblast clones leads to tumor-like overgrowth through both a severe disruption of the asymmetric cell division process and a canoe loss-mediated Ras-PI3K-Akt activation. Moreover, canoe loss also interacts synergistically with scribble to promote overgrowth in epithelial tissues, here just by activating the Ras-Raf-MAPK pathway. Finally scribble functionally related genes discs large and lethal (2) giant larvae were shown to contribute to repress the Ras-MAPK signaling cascade in epithelia. Hence, this work uncovers novel cooperative interactions between all these well-conserved tumor suppressors to ensure a tight regulation of the Ras signaling pathway.
Rui, M., Qian, J., Liu, L., Cai, Y., Lv, H., Han, J., Jia, Z. and Xie, W. (2017). The neuronal protein Neurexin directly interacts with the Scribble-Pix complex to stimulate F-actin assembly for synaptic vesicle clustering. J Biol Chem [Epub ahead of print]. PubMed ID: 28710284
Summary:
Synaptic vesicles (SVs) form distinct pools at synaptic terminals, and this well-regulated separation is necessary for normal neuro-transmission. However, how SV cluster in particular synaptic compartments to maintain normal neurotransmitter release remains a mystery. The presynaptic protein Neurexin (NRX) plays a significant role in synaptic architecture and function, and some evidences suggest that NRX is associated with neurological disorders, including autism spectrum disorders. However, the role of NRX in SV clustering is unclear. Using the neuromuscular junction at the 2-3 instar stages of Drosophila larvae as a model and biochemical, imaging, and electrophysiology techniques, this study demonstrate that Drosophila NRX (DNRX) plays critical roles in regulating synaptic terminal clustering and release of SVs. DNRX controls the terminal clustering and release of SVs by stimulating presynaptic F-actin. Furthermore, the results indicate that DNRX functions through the scaffold protein Scribble and the GEF protein Pixie to activate the small GTPase Rac1. A direct interaction was observed between the C-terminal PDZ-binding motif of DNRX and the PDZ domains of Scribble and that Scribble bridges DNRX to DPix, forming a DNRX/Scribble/DPix complex that activates Rac1 and subsequently stimulates presynaptic F-actin assembly and SV clustering. Taken together, this work provides important insights into the function of DNRX in regulating SV clustering, which could help inform further research into pathological neurexin-mediated mechanisms in neurological disorders such as autism.
Schnaitmann, C., Haikala, V., Abraham, E., Oberhauser, V., Thestrup, T., Griesbeck, O. and Reiff, D. F. (2018). Color processing in the early visual system of Drosophila. Cell 172(1-2): 318-330.e318. PubMed ID: 29328919
Summary:
Color vision is encoded by color-opponent neurons that are excited at one wavelength and inhibited at another. This study examined the circuit implementation of color-opponent processing in the Drosophila visual system by combining two-photon calcium imaging with genetic dissection of visual circuits. Color-opponent processing of UVshort/blue and UVlong/green is already implemented in R7/R8 inner photoreceptor terminals of "pale" and "yellow" ommatidia, respectively. R7 and R8 photoreceptors of the same type of ommatidia mutually inhibit each other directly via HisCl1 histamine receptors and receive additional feedback inhibition that requires the second histamine receptor Ort. Color-opponent processing at the first visual synapse represents an unexpected commonality between Drosophila and vertebrates; however, the differences in the molecular and cellular implementation suggest that the same principles evolved independently.
Caria, S., Magtoto, C. M., Samiei, T., Portela, M., Lim, K. Y. B., How, J. Y., Stewart, B. Z., Humbert, P. O., Richardson, H. E. and Kvansakul, M. (2018). Drosophila melanogaster Guk-holder interacts with the Scribbled PDZ1 domain and regulates epithelial development with Scribbled and Discs Large. J Biol Chem [Epub ahead of print]. PubMed ID: 29378849
Summary:
Epithelial cell polarity is controlled by components of the Scribble polarity module, and its regulation is critical for tissue architecture and cell proliferation and cell migration. In Drosophila melanogaster, the adaptor protein Guk-holder (Gukh) binds to the Scribbled (Scrib) and Discs Large (Dlg) components of the Scribble polarity module and plays an important role in the formation of neuromuscular junctions. However, Gukhs role in epithelial tissue formation and the molecular basis for the Scrib-Gukh interaction remain to be defined. This study shows using isothermal titration calorimetry that the Scrib PDZ1 domain is the major site for an interaction with Gukh. Furthermore, the structural basis of this interaction is defined by determining the crystal structure of the Scrib PDZ1-Gukh complex. The C-terminal PDZ-binding motif of Gukh is located in the canonical ligand binding groove of Scrib PDZ1, and utilizes an unusually extensive network of hydrogen bonds and ionic interactions to enable binding to PDZ1 with high affinity. The roles of Gukh along with those of Scrib and Dlg were examined in Drosophila epithelial tissues, and found Gukh was found to be expressed in larval-wing and eye-epithelial tissues and co-localizes with Scrib and Dlg at the apical cell cortex. Importantly, it was shown that Gukh functions with Scrib and Dlg in the development of Drosophila epithelial tissues, with depletion of Gukh enhancing the eye- and wing-tissue defects caused by Scrib or Dlg depletion. Overall these findings reveal that Scribs PDZ1 domain functions in the interaction with Gukh and that the Scrib-Gukh interaction has a key role in epithelial tissue development in Drosophila.
La Fortezza, M., Grigolon, G., Cosolo, A., Pinduyrin, A., Breimann, L., Blum, H., van Steensel, B. and Classen, A. K. (2018). DamID profiling of dynamic Polycomb-binding sites in Drosophila imaginal disc development and tumorigenesis. Epigenetics Chromatin 11(1): 27. PubMed ID: 29871666
Summary:
Tracking dynamic protein-chromatin interactions in vivo is key to unravel transcriptional and epigenetic transitions in development and disease. However, limited availability and heterogeneous tissue composition of in vivo source material impose challenges on many experimental approaches. This study has adapted cell-type-specific DamID-seq profiling for use in Drosophila imaginal discs and make FLP/FRT-based induction accessible to GAL driver-mediated targeting of specific cell lineages. In a proof-of-principle approach, ubiquitous DamID expression was used to describe dynamic transitions of Polycomb-binding sites during wing imaginal disc development and in a scrib tumorigenesis model. Atf3 and Ets21C as novel Polycomb target genes involved in scrib tumorigenesis and suggest that target gene regulation by Atf3 and AP-1 transcription factors, as well as modulation of insulator function, plays crucial roles in dynamic Polycomb-binding at target sites. These findings by DamID-seq analysis of wing imaginal disc samples derived from 10 larvae. This study opens avenues for robust profiling of small cell population in imaginal discs in vivo and provides insights into epigenetic changes underlying transcriptional responses to tumorigenic transformation.
Strassburger, K., Kang, E. and Teleman, A. A. (2019). Drosophila ZDHHC8 palmitoylates scribble and Ras64B and controls growth and viability. PLoS One 14(2): e0198149. PubMed ID: 30735487
Summary:
Palmitoylation is an important posttranslational modification regulating diverse cellular functions. Consequently, aberrant palmitoylation can lead to diseases such as neuronal disorders or cancer. In humans there are roughly one hundred times more palmitoylated proteins than enzymes catalyzing palmitoylation (palmitoyltransferases). Therefore, it is an important challenge to establish the links between palmitoyltransferases and their targets. From publicly available data, this study found that expression of human ZDHHC8 correlates significantly with cancer survival. To elucidate the organismal function of ZDHHC8, the Drosophila ortholog of hZDHHC8, CG34449/dZDHHC8 was studied. Knockdown of dZDHHC8 causes tissue overgrowth while dZDHHC8 mutants are larval lethal. A list is provided of 159 palmitoylated proteins in Drosophila and data is presented suggesting that Scribble and Ras64B are targets of dZDHHC8.
Nakajima, Y. I., Lee, Z. T., McKinney, S. A., Swanson, S. K., Florens, L. and Gibson, M. C. (2019). Junctional tumor suppressors interact with 14-3-3 proteins to control planar spindle alignment. J Cell Biol. PubMed ID: 31088859
Summary:
Proper orientation of the mitotic spindle is essential for cell fate determination, tissue morphogenesis, and homeostasis. During epithelial proliferation, planar spindle alignment ensures the maintenance of polarized tissue architecture, and aberrant spindle orientation can disrupt epithelial integrity. Nevertheless, in vivo mechanisms that restrict the mitotic spindle to the plane of the epithelium remain poorly understood. This study shows that the junction-localized tumor suppressors Scribbled (Scrib) and Discs large (Dlg) control planar spindle orientation via Mud and 14-3-3 proteins in the Drosophila wing disc epithelium. During mitosis, Scrib is required for the junctional localization of Dlg, and both affect mitotic spindle movements. Using coimmunoprecipitation and mass spectrometry, this study identified 14-3-3 proteins as Dlg-interacting partners and further reports that loss of 14-3-3s causes both abnormal spindle orientation and disruption of epithelial architecture as a consequence of basal cell delamination and apoptosis. Combined, these biochemical and genetic analyses indicate that 14-3-3s function together with Scrib, Dlg, and Mud during planar cell division.
Sharifkhodaei, Z., Gilbert, M. M. and Auld, V. J. (2019). Scribble and Discs-large mediate tricellular junction formation. Development. PubMed ID: 31444218
Summary:
Junctional complexes that mediate cell adhesion are key to epithelial integrity, cell division and permeability barriers formation. In Drosophila the scaffolding proteins Scribble (Scrib) and Discs-large (Dlg) are key regulators of epithelial polarity, proliferation, assembly of junctions, and protein trafficking. This study found that Scrib and Dlg are necessary for the formation of the tricellular junction (TCJ), a unique junction that forms in epithelia at the convergence of three neighbouring cells. Scrib and Dlg are in close proximity with the TCJ proteins Gliotactin (Gli) and Bark-beetle (Bark) and both are required for TCJ protein recruitment. Loss of Bark or Gli lead to the basolateral spread of the TCJ complex at the cell corners. Loss of the septate junction proteins NrxIV and the Na(+)/K(+) ATPase also resulted in the basolateral spread of the entire TCJ complex at the cell corners. The Scrib PDZ1-2 domains and the Dlg GUK domain are necessary for Bark and Gli localization to the TCJ. Overall, a model is proposed where Scrib and Dlg are key components of the TCJ, and form a complex with Bark and Gli.
Zhang, W., Reeves, R. G. and Tautz, D. (2019). Identification of a genetic network for an ecologically relevant behavioral phenotype in Drosophila melanogaster. Mol Ecol. PubMed ID: 31867742
Summary:
Pupation site choice of Drosophila third-instar larvae is critical for the survival of individuals, as pupae are exposed to various biotic and abiotic dangers while immobilized during the 3-4 days of metamorphosis. This singular behavioural choice is sensitive to both environmental and genetic factors. This study developed a high-throughput phenotyping approach to assay the variation in pupation height in Drosophila melanogaster, while controlling for possibly confounding factors. Substantial variation was found of mean pupation height among sampled natural stocks, and it was shown that the Drosophila Genetic Reference Panel (DGRP) reflects this variation. Using the DGRP stocks for genome wide association (GWA) mapping, 16 loci involved in determining pupation height could be resolved. The candidate genes in these loci are enriched for high expression in the larval central nervous system. A genetic network could be constructed from the candidate loci, which places scribble (scrib) at the centre, plus other genes known to be involved in nervous system development, such as Epidermal growth factor receptor (Egfr) and p53. Using gene disruption lines, it was possible to functionally validate several of the initially identified loci, as well as additional loci predicted from network analysis. This study shows that the combination of high throughput phenotyping with a genetic analysis of variation captured from the wild can be used to approach the genetic dissection of an environmentally relevant behavioural phenotype.
Wei, T., Ji, X., Gao, Y., Zhu, X. and Xiao, G. (2021). ZnT7 RNAi favors Raf(GOF)scrib(-/-)-induced tumor growth and invasion in Drosophila through JNK signaling pathway. Oncogene. PubMed ID: 33649534
Summary:
The disruption of zinc homeostasis has been identified in patients suffering from various cancers, but a causative relationship has not yet been established. Drosophila melanogaster has become a powerful model to study cancer biology. Using a Drosophila model of malignant tumor RafGOFscrib-/-, it was observed that the tumor growth, invasion and migration were enhanced by silencing dZnT7, a zinc transporter localized on the Golgi apparatus. Further study indicated that the zinc deficiency in Golgi of dZnT7 RNAi resulted in ER stress which could activate the c-Jun-N-terminal Kinase (JNK) signaling and this process is mediated by Atg9. Lastly, it was demonstrated that the exacerbation of dZnT7 RNAi on tumor was promoted by JNK signaling-dependent cell autonomous and non-autonomous autophagy. These findings suggest that zinc homeostasis in secretory compartments may provide a new therapeutic target for tumor treatment.
Kohashi, K., Mori, Y., Narumi, R., Kozawa, K., Kamasaki, T., Ishikawa, S., Kajita, M., Kobayashi, R., Tamori, Y. and Fujita, Y. (2021). Sequential oncogenic mutations influence cell competition. Curr Biol. PubMed ID: 34314674
Summary:
At the initial stage of carcinogenesis, newly emerging transformed cells are often eliminated from epithelial layers via cell competition with the surrounding normal cells. For instance, when surrounded by normal cells, oncoprotein RasV12-transformed cells are extruded into the apical lumen of epithelia. During cancer development, multiple oncogenic mutations accumulate within epithelial tissues. However, it remains elusive whether and how cell competition is also involved in this process. Using a mammalian cell culture model system, this study investigated what happens upon the consecutive mutations of Ras and tumor suppressor protein Scribble. When Ras mutation occurs under the Scribble-knockdown background, apical extrusion of Scribble/Ras double-mutant cells is strongly diminished. In addition, at the boundary with Scribble/Ras cells, Scribble-knockdown cells frequently undergo apoptosis and are actively engulfed by the neighboring Scribble/Ras cells. The comparable apoptosis and engulfment phenotypes are also observed in Drosophila epithelial tissues between Scribble/Ras double-mutant and Scribble single-mutant cells. Furthermore, mitochondrial membrane potential is enhanced in Scribble/Ras cells, causing the increased mitochondrial reactive oxygen species (ROS). Suppression of mitochondrial membrane potential or ROS production diminishes apoptosis and engulfment of the surrounding Scribble-knockdown cells, indicating that mitochondrial metabolism plays a key role in the competitive interaction between double- and single-mutant cells. Moreover, mTOR (mechanistic target of rapamycin kinase) acts downstream of these processes. These results imply that sequential oncogenic mutations can profoundly influence cell competition, a transition from loser to winner. Further studies would open new avenues for cell competition-based cancer treatment, thereby blocking clonal expansion of more malignant populations within tumors.
Cong, B., Nakamura, M., Sando, Y., Kondo, T., Ohsawa, S. and Igaki, T. (2021). JNK and Yorkie drive tumor malignancy by inducing L-amino acid transporter 1 in Drosophila. PLoS Genet 17(11): e1009893. PubMed ID: 34780467.
Summary:
Identifying a common oncogenesis pathway among tumors with different oncogenic mutations is critical for developing anti-cancer strategies. This study performed transcriptome analyses on two different models of Drosophila malignant tumors caused by Ras activation with cell polarity defects (RasV12/scrib-/-) or by microRNA bantam overexpression with endocytic defects (bantam/rab5-/-), followed by an RNAi screen for genes commonly essential for tumor growth and malignancy. Juvenile hormone Inducible-21 (JhI-21), a Drosophila homolog of the L-amino acid transporter 1 (LAT1), was identified is upregulated in these malignant tumors with different oncogenic mutations and knocking down of JhI-21 strongly blocked their growth and invasion. JhI-21 expression was induced by simultaneous activation of c-Jun N-terminal kinase (JNK) and Yorkie (Yki) in these tumors and thereby contributed to tumor growth and progression by activating the mTOR-S6 pathway. Pharmacological inhibition of LAT1 activity in Drosophila larvae significantly suppressed growth of RasV12/scrib-/- tumors. Intriguingly, LAT1 inhibitory drugs did not suppress growth of bantam/rab5-/- tumors and overexpression of bantam rendered RasV12/scrib-/- tumors unresponsive to LAT1 inhibitors. Further analyses with RNA sequencing of bantam-expressing clones followed by an RNAi screen suggested that bantam induces drug resistance against LAT1 inhibitors via downregulation of the TMEM135-like gene CG31157. These observations unveil an evolutionarily conserved role of LAT1 induction in driving Drosophila tumor malignancy and provide a powerful genetic model for studying cancer progression and drug resistance.
Manzanero-Ortiz, S., de Torres-Jurado, A., Hernandez-Rojas, R. and Carmena, A. (2021). Pilot RNAi Screen in Drosophila Neural Stem Cell Lineages to Identify Novel Tumor Suppressor Genes Involved in Asymmetric Cell Division. Int J Mol Sci 22(21). PubMed ID: 34768763
Summary:
A connection between compromised asymmetric cell division (ACD) and tumorigenesis was proven some years ago using Drosophila larval brain neural stem cells, called neuroblasts (NBs), as a model system. Since then, it has been learned that compromised ACD does not always promote tumorigenesis, as ACD is an extremely well-regulated process in which redundancy substantially overcomes potential ACD failures. Considering this, a pilot RNAi screen was performed in Drosophila larval brain NB lineages using Ras(V)(12) scribble (scrib) mutant clones as a sensitized genetic background, in which ACD is affected but does not cause tumoral growth. First, as a proof of concept, this study has tested known ACD regulators in this sensitized background, such as lethal (2) giant larvae and warts. Although the downregulation of these ACD modulators in NB clones does not induce tumorigenesis, their downregulation along with Ras(V)(12) scrib does cause tumor-like overgrowth. Based on these results, 79 RNAi lines randomly screened detecting 15 potential novel ACD regulators/tumor suppressor genes. It is concluded that Ras(V)(12) scrib is a good sensitized genetic background in which to identify tumor suppressor genes involved in NB ACD, whose function could otherwise be masked by the high redundancy of the ACD process.
Logeay, R., Geminard, C., Lassus, P., Rodriguez-Vazquez, M., Kantar, D., Heron-Milhavet, L., Fischer, B., Bray, S. J., Colinge, J. and Djiane, A. (2022). Mechanisms underlying the cooperation between loss of epithelial polarity and Notch signaling during neoplastic growth in Drosophila. Development. PubMed ID: 35005772
Summary:
Aggressive neoplastic growth can be initiated by a limited number of genetic alterations, such as the well-established cooperation between loss of cell architecture and hyperactive signaling pathways. However, understanding of how these different alterations interact and influence each other remains very incomplete. Using Drosophila paradigms of imaginal wing disc epithelial growth, this study monitored the changes in Notch pathway activity according to the polarity status of cells (scrib mutant). The scrib mutation was shown to impact the direct transcriptional output of the Notch pathway, without altering the global distribution of Su(H), the Notch dedicated transcription factor. The Notch-dependent neoplasms require however, the action of a group of transcription factors, similar to those previously identified for Ras/scrib neoplasm (namely AP-1, Stat92E, Ftz-F1, and bZIP factors), further suggesting the importance of this transcription factor network during neoplastic growth. Finally this work highlights some Notch/scrib specificities, in particular the role of the PAR domain containing bZIP transcription factor and Notch direct target Pdp1 for neoplastic growth.
Khoury, M. J. and Bilder, D. (2022). Minimal functional domains of the core polarity regulator Dlg Biol Open. PubMed ID: 35722710
Summary:
The compartmentalized domains of polarized epithelial cells arise from mutually antagonistic actions between the apical Par complex and the basolateral Scrib module. In Drosophila, the Scrib module proteins Scribble (Scrib) and Discs-large (Dlg) are required to limit Lgl phosphorylation at the basolateral cortex, but how Scrib and Dlg could carry out such a 'protection' activity is not clear. This study tested Protein Phosphatase 1α (PP1) as a potential mediator of this activity but demonstrate that a significant component of Scrib and Dlg regulation of Lgl is PP1-independent, and found no evidence for a Scrib-Dlg-PP1 protein complex. However, the Dlg SH3 domain plays a role in Lgl protection and, in combination with the N-terminal region of the Dlg HOOK domain, in recruitment of Scrib to the membrane. A 'minimal Dlg' was identified, comprised of the SH3 and HOOK domains that is both necessary and sufficient for Scrib localization and epithelial polarity function in vivo.
Gerlach, S. U., de Vreede, G. and Bilder, D. (2022). PTP10D-mediated cell competition is not obligately required for elimination of polarity-deficient clones. Biol Open 11(11). PubMed ID: 36355597
Summary:
Animal organs maintain tissue integrity and ensure removal of aberrant cells through several types of surveillance mechanisms. One prominent example is the elimination of polarity-deficient mutant cells within developing Drosophila imaginal discs. This has been proposed to require heterotypic cell competition dependent on the receptor tyrosine phosphatase PTP10D within the mutant cells. This study reports experiments to test this requirement in various contexts and found that PTP10D is not obligately required for the removal of scribble (scrib) mutant and similar polarity-deficient cells. These experiments used identical stocks with which another group can detect the PTP10D requirement, and the results do not vary under several husbandry conditions including high and low protein food diets. Although it was not possible to identify the source of the discrepant results, it is suggested that the role of PTP10D in polarity-deficient cell elimination may not be absolute.
Worley, M. I., Everetts, N. J., Yasutomi, R., Chang, R. J., Saretha, S., Yosef, N. and Hariharan, I. K. (2022). Ets21C sustains a pro-regenerative transcriptional program in blastema cells of Drosophila imaginal discs. Curr Biol 32(15): 3350-3364. PubMed ID: 35820420
Summary:
An important unanswered question in regenerative biology is to what extent regeneration is accomplished by the reactivation of gene regulatory networks used during development versus the activation of regeneration-specific transcriptional programs. Following damage, Drosophila imaginal discs, the larval precursors of adult structures, can regenerate missing portions by localized proliferation of damage-adjacent tissue. Using single-cell transcriptomics in regenerating wing discs, a comprehensive view of the transcriptome of regenerating discs was obtained and two regeneration-specific cell populations within the blastema, Blastema1 and Blastema2 were identified. Collectively, these cells upregulate multiple genes encoding secreted proteins that promote regeneration including Pvf1, upd3, asperous, Mmp1, and the maturation delaying factor Ilp8. Expression of the transcription factor Ets21C is restricted to this regenerative secretory zone; it is not expressed in undamaged discs. Ets21C expression is activated by the JNK/AP-1 pathway, and it can function in a type 1 coherent feedforward loop with AP-1 to sustain expression of downstream genes. Without Ets21C function, the blastema cells fail to maintain the expression of a number of genes, which leads to premature differentiation and severely compromised regeneration. As Ets21C is dispensable for normal development, these observations indicate that Ets21C orchestrates a regeneration-specific gene regulatory network. This study has also identified cells resembling both Blastema1 and Blastema2 in scribble tumorous discs. They express the Ets21C-dependent gene regulatory network, and eliminating Ets21C function reduces tumorous growth. Thus, mechanisms that function during regeneration can be co-opted by tumors to promote aberrant growth.
Gerlach, S. U., de Vreede, G. and Bilder, D. (2022). PTP10D-mediated cell competition is not obligately required for elimination of polarity-deficient clones. Biol Open 11(11). PubMed ID: 36355597
Summary:
Animal organs maintain tissue integrity and ensure removal of aberrant cells through several types of surveillance mechanisms. One prominent example is the elimination of polarity-deficient mutant cells within developing Drosophila imaginal discs. This has been proposed to require heterotypic cell competition dependent on the receptor tyrosine phosphatase PTP10D within the mutant cells. This study reports experiments to test this requirement in various contexts and found that PTP10D is not obligately required for the removal of scribble (scrib) mutant and similar polarity-deficient cells. These experiments used identical stocks with which another group can detect the PTP10D requirement, and the results do not vary under several husbandry conditions including high and low protein food diets. Although it was not possible to identify the source of the discrepant results, it is suggested that the role of PTP10D in polarity-deficient cell elimination may not be absolute.
La Marca, J. E., Ely, R. W., Diepstraten, S. T., Burke, P., Kelly, G. L., Humbert, P. O. and Richardson, H. E. (2023). A Drosophila chemical screen reveals synergistic effect of MEK and DGKα inhibition in Ras-driven cancer. Dis Model Mech 16(3). PubMed ID: 36861754
Summary:
Elevated Ras signalling is highly prevalent in human cancer; however, targeting Ras-driven cancers with Ras pathway inhibitors often leads to undesirable side effects and to drug resistance. Thus, identifying compounds that synergise with Ras pathway inhibitors would enable lower doses of the Ras pathway inhibitors to be used and also decrease the acquisition of drug resistance. Here, in a specialised chemical screen using a Drosophila model of Ras-driven cancer, this study has identified compounds that reduce tumour size by synergising with sub-therapeutic doses of the Ras pathway inhibitor trametinib, which targets MEK, the mitogen-activated protein kinase kinase, in this pathway. Analysis of one of the hits, ritanserin, and related compounds revealed that diacyl glycerol kinase α (DGKα, Dgk in Drosophila) was the critical target required for synergism with trametinib. Human epithelial cells harbouring the H-RAS oncogene and knockdown of the cell polarity gene SCRIB were also sensitive to treatment with trametinib and DGKα inhibitors. Mechanistically, DGKα inhibition synergises with trametinib by increasing the P38 stress-response signalling pathway in H-RASG12V SCRIBRNAi cells, which could lead to cell quiescence. These results reveal that targeting Ras-driven human cancers with Ras pathway and DGKα inhibitors should be an effective combination drug therapy.
Song, F., Zhang, W., Li, X., Chen, X., Yuan, X., Jiang, M., Zhao, Y., Liu, Q. and Zhou, Z. (2023). FSBP suppresses tumor cell migration by inhibiting the JNK pathway. iScience 26(4): 106440. PubMed ID: 37035004
Summary:
The main cause of high mortality in cancer patients is tumor metastasis. Exploring the underlying mechanism of tumor metastasis is of great significance for clinical treatments. This study has identified the transcription factor Apt/FSBP is a suppressor for tumor metastasis. In Drosophila wing disc, knockdown of apt is able to trigger cell migration, whereas overexpression of apt hampers scrib-RNAi-induced tumor cell migration. Further studies show that loss of apt promotes cell migration JNK pathway . To investigate the role of FSBP, the homolog of Apt in mammals, Fsbp liver-specific knockout mice were constructed. Knockout of Fsbp in liver does not cause any detectable physiological defects, but predisposes to tumorigenesis on DEN and CCl(4) treatment. In addition, loss of Fsbp accelerates tumor metastasis from liver to diaphragm. Taken together, this study uncovers FSBP is a novel tumor suppressor, and provides it as a considerable drug target for tumor treatment.

BIOLOGICAL OVERVIEW

Loss of cell polarity and tissue architecture are characteristics of malignant cancers derived from epithelial tissues. Cells in epithelial sheets are characterized by columnar or cuboidal shape, strong cell-cell adhesion, and pronounced apicobasal polarity. However, tumors of epithelial origin lose these characteristics as they progress from benign growth to malignant carcinoma, and this loss is associated with poor clinical prognosis. Evidence is provided that a group of membrane-associated proteins act in concert to regulate both epithelial structure and cell proliferation. Scribbled (Scrib) is a cell junction-localized protein required for polarization of embryonic and imaginal disc and follicular epithelia. The tumor suppressor scrib was isolated in a screen for maternal effect mutations that disrupt aspects of epithelial morphogenesis such as cell adhesion, shape and polarity. Commonly referred to as 'scribble' both here and in the literature, the gene name is maintained as 'scribbled' by FlyBase, to avoid (or enhance) confusion. scrib encodes a multi-PDZ (PSD-95, Discs-large and ZO-1) and leucine-rich-repeat protein. The structure of the embryonic cuticle was used to reflect the organization of the underlying epithelial epidermis that secretes it. The wild-type cuticle forms a smooth, continuous sheet, but embryos that are maternally and zygotically mutant for scrib produce a corrugated cuticular surface that is riddled with holes, hence the name scribbled (Bilder, 2000a).

To place Scrib within the known pathway for Drosophila epithelial polarity determination, the effect of scrib mutations on Crumbs (Crb) was examined. Crb is an apically localized transmembrane protein that is necessary and sufficient to confer apical character on plasma membrane. In scrib embryos, Crb shows unrestricted localization in both apical and basolateral regions. Whether scrib mutants are identical to a gain-of-function crb phenotype was examined by comparing them with embryos in which GAL4-driven Crb is present throughout the cell membrane. Ectopic Crb that is produced in this manner is sufficient to phenocopy several aspects of scrib embryos, including mislocalization of apical proteins and the cuticle pattern. These data indicate that a major function of Scrib in epithelial polarity is to exclude Crb from the basolateral domain. Since ectopic Crb does not cause the epithelial morphology and multilayering defects seen in scrib embryos, Scrib may be required for the localization of additional epithelial determinants as well (Bilder, 2000a).

Analysis of the morphological and polarization phenotypes exhibited by scrib embryos shows that Scrib is a critical component of epithelial architecture in the Drosophila ectoderm, and suggests that its function is closely linked to that of Crb. Scrib is not required for the early localization of basal Discs lost (now redefined as Drosophila Patj) or apical Armadillo during blastoderm formation, and scrib mutants do not exhibit the defective cellularization or precipitous loss of cell adhesion seen when Discs lost or Armadillo, respectively, are depleted in the embryo. The increasingly severe cell shape, polarity and epithelial organization defects of scrib embryos are first manifested after gastrulation, coincident with the onset of defects in crb and stardust embryos. Loss of Crb results in loss of apical proteins from the plasma membrane and a failure to consolidate early adherens junction material into an apical band of zonula adherins (ZAs), while in scrib embryos early adherens junctions become misdistributed basolaterally. Together with the similarities between scrib loss-of-function and crb gain-of-function phenotypes, these data place Scrib and Crb in a pathway required for the progression from the initially differentiated blastoderm membrane domains into a fully polarized epithelium with mature junctions (Bilder, 2000a).

These results show that the junctional protein Scrib specifically restricts apical membrane determinants to the apical cell surface. This restriction allows the proper segregation of apical and basolateral domain components, and the appropriate placement of the adherens junction, resulting in full epithelial polarization. How does Scrib, a putative scaffolding protein whose localizaton bounds the apical domain, dictate the proper confinement of apical proteins? Two models suggest themselves. Scrib could assemble a diffusion barrier that physically separates apical and basolateral compartments, similar to the 'fence' function proposed for the vertebrate tight junction. To date, such a barrier has been shown to exist only for lipid diffusion in the outer leaflet of the plasma membrane. If scrib mutations disrupt such a mechanical barrier, then secondary retention systems must serve to maintain basolateral protein restriction from the apical cell surface. An alternative is that Scrib has a role in the polarized targeting of transport vesicles carrying apical proteins. The junctional complex, and in particular the tight junction, has been proposed to be a key sorting site for a subset of Golgi-derived vesicles. In this model, Scrib might interact with the 'exocyst', a secretory targeting apparatus localized to the tight junction and involved in polarized segregation of transmembrane proteins. PDZ domain proteins have been implicated at several different sites of the protein trafficking pathway, and occasional punctate intracellular staining of Scrib is reminiscent of vesicles. Distinction between these models will rely on the identification of binding partners for Scrib (Bilder, 2000a).

Two other tumor suppressors, lethal giant larvae (lgl) and discs-large (dlg), have the identical effects as scrib mutation on epithelial structure. Scrib and Dlg colocalize and overlap with Lgl in epithelia; activity of all three genes is required for cortical localization of Lgl and junctional localization of Scrib and Dlg. scrib, dlg, and lgl show strong genetic interactions. Thus, these three tumor suppressors act together in a common pathway to regulate cell polarity and growth control (Bilder, 2000b).

To test whether the requirement for Scrib is limited to the embryo, the role of scrib in follicle cells, a monolayered epithelium of somatic cells that encases the germ line in the adult female ovary, was examined. Scrib is localized to lateral follicle cell membranes, and clones of cells that lack scrib function become round and multilayered with polarity defects, similar to the phenotype of embryonic epithelia lacking scrib function. The epithelial defects of scrib mutant follicle cells are cell autonomous. These data indicate that scrib is required within cells from multiple tissues for proper epithelial structure (Bilder, 2000b).

Because follicle cell epithelia require scrib, lgl, and dlg, the functions of lgl and dlg were examined in the embryonic epidermis, where scrib acts to restrict apical proteins and adherens junctions to their appropriate positions within the cell membrane (Bilder, 2000a). Embryos lacking both maternal and zygotic contributions of lgl and dlg, (hereafter referred to as lgl and dlg embryos) were stained with antibodies to polarized proteins and cellular junction components. During mid-embryogenesis, lgl and dlg embryos show defects in apicobasal polarity, revealed by aberrant distribution of the apical protein Crumbs (Crb) and disruption of adherens junctions. These defects are similar to those of scrib embryos; the terminal phenotypes of scrib, lgl, and dlg embryos, as indicated by cuticle deposition, are also nearly identical. Thus, lgl and dlg, like scrib, act to properly localize apical proteins and adherens junctions to organize epithelial architecture in embryos (Bilder, 2000b).

The similarity of mutant phenotypes in different epithelia suggests that the three proteins are components of the fundamental machinery that creates the distinctive architecture of epithelial cells and tissues. To test this assertion, the scrib phenotype was compared to that of lgl and dlg in a third major epithelium, the larval imaginal disc. Discs isolated from late third instar larvae zygotically mutant for scrib are profoundly disorganized and also massively overgrown. scrib discs contain 4.7 times as many cells as wild-type (WT) discs and consist of spherical masses of tightly packed cells, as opposed to the folded monolayer epithelium seen in WT larvae. The apical polarization of actin evident in WT discs is absent in scrib discs. This loss of epithelial organization accompanied by overproliferation corresponds to the phenotype described for lgl and dlg zygotic mutant discs. Additional features of lgl and dlg larval phenotypes, such as overgrowth of brain tissue, are also present in scrib larvae. Together, these data indicate that scrib and the two previously characterized Drosophila malignant neoplastic tumor suppressors, lgl and dlg, share a role in growth control as well as epithelial polarity. Epistatic relations between scrib, lgl, and dlg were investigated by determining the localization of each protein in embryos mutant for the other two genes. These experiments have shown that dlg is required for the stable association of Scrib with the cell membrane and scrib is required for the cortical association of Lgl; all three genes act to localize Scrib and Dlg to the apical margin of the lateral membrane (ALM) of the embryonic epidermal epithelium (Bilder, 2000b).

These results provide strong evidence that Scrib, Dlg, and Lgl act in a common pathway to regulate cell architecture and cell proliferation control. Of the ~50 Drosophila genes in which mutation gives rise to overproliferation, only scrib shares with dlg and lgl the concomitant loss of tissue organization that groups the three together as malignant neoplastic tumor suppressors. Previous analyses have described a role for dlg and lgl in imaginal disc polarity; the demonstration in this work of genetic interactions with scrib and codependence for protein localization indicates a functional link between the three tumor suppressors. Furthermore, involvement of the tumor suppressors in embryonic epithelial polarity provides a well-studied context in which to understand their activities. These findings suggest that, in the WT gastrula, intrinsic, perhaps adhesion-based cues localize Dlg at the ALM; Dlg stabilizes Scrib at this position, and finally Scrib acts on the cortical cytoskeleton to bring Lgl to the membrane. The three proteins may then collaborate to maintain the proper distribution of polarized factors, including themselves (Bilder, 2000b).

The correlation between loss of membrane-associated Lgl in scrib and dlg mutants and defective cell polarity suggests models of action for this group of proteins. Whereas the PDZ domains of Scrib and Dlg are likely to bind to transmembrane proteins that organize the epithelial cell surface, the role of Lgl in polarity determination may derive from its function in targeted secretion of membrane proteins. Lgl homologs from humans and yeast can bind to plasma membrane t-SNARE proteins and promote the fusion of cargo-carrying vesicles with target membranes (Fujita, 1998; Lehman, 1999). In yeast undergoing polarized growth, the broadly distributed Lgl homologs function primarily at the bud tip, the site of the 'exocyst' complex required for vesicle trafficking and addition (TerBush, 1996). In vertebrate epithelia, exocyst components are found at the tight junction, a structure analogous to the septate junction where Dlg and Scrib localize. In Drosophila epithelia, recruitment of Lgl into the proximity of membrane t-SNAREs requires proper localization of Scrib and Dlg, thus potentially linking the transmembrane proteins that establish polarity to the protein-targeting system that preserves it (Bilder, 2000b and references therein).

In many epithelial-derived cancers, cytoarchitectural changes are hallmarks of oncogenic transformation. The disruption of epithelial architecture seen in scrib, dlg, and lgl animals could affect growth control by several mechanisms. Many growth factor receptors are polarized to a specific membrane domain, and mislocalization of such proteins may affect signaling pathways that maintain cells in a differentiated, nonproliferative state. Additionally, the aberrant cell-cell junctions formed in scrib, dlg, and lgl mutants could compromise contact inhibition. Finally, disruption of cell-cell contacts may release junction-localized signaling components, such as Arm or APC, that have been implicated in regulating cell proliferation; indeed, a human Dlg homolog has been shown to bind APC and associate with beta-catenin, the human homolog of Arm. Because the modes of action of Scrib, Dlg, and Lgl are likely to be conserved between vertebrates and invertebrates, investigation into a tumorigenic role for the multiple human homologs of these genes is warranted. Further analysis of the mechanisms by which Scrib, Dlg, and Lgl keep Drosophila cell growth in check will likely enhance an understanding of mammalian oncogenesis as well (Bilder, 2000b).

Fat body-derived Spz5 remotely facilitates tumor-suppressive cell competition through Toll-6-α-Spectrin axis-mediated Hippo activation

Tumor-suppressive cell competition is an evolutionarily conserved process that selectively removes precancerous cells to maintain tissue homeostasis. Using the polarity-deficiency-induced cell competition model in Drosophila, this study identify Toll-6, a Toll-like receptor family member, as a driver of tension-mediated cell competition through α-Spectrin (α-Spec)-Yorkie (Yki) cascade. Toll-6 aggregates along the boundary between wild-type and polarity-deficient clones, where Toll-6 physically interacts with the cytoskeleton network protein α-Spec to increase mechanical tension, resulting in actomyosin-dependent Hippo pathway activation and the elimination of scrib mutant cells. Furthermore, this study show that Spz5 secreted from fat body, the key innate organ in fly, facilitates the elimination of scrib clones by binding to Toll-6. These findings uncover mechanisms by which fat bodies remotely regulate tumor-suppressive cell competition of polarity-deficient tumors through inter-organ crosstalk and identified the Toll-6-α-Spec axis as an essential guardian that prevents tumorigenesis via tension-mediated cell elimination (Kong, 2022).

Epithelial cells possess intrinsic mechanisms to outcompete and eliminate early precancerous cells to maintain homeostasis. For instance, in a mouse model of esophageal carcinogenesis, the majority of newly developed tumor clones are eliminated through cell competition by adjacent normal epithelium. Similarly, surveillance mechanisms also exist in Drosophila epithelium to actively remove oncogenic clones composed of polarity-deficient cells. Genetic studies in flies have uncovered numerous mechanisms that regulate tumor-suppressive cell competition, including c-Jun-N-terminal kinase (JNK) signaling activation-mediated cell elimination, direct cell-cell interaction, secreted factors from epithelial cells, and inter-organ crosstalk between insulin-producing cells and precancerous cell-bearing discs (Kong, 2022).

Initially identified in Drosophila, the Hippo pathway is an evolutionarily conserved signaling cascade that plays crucial roles in various physiological and pathological contexts, ranging from tumor progression and embryogenesis to stem cell renewal and immune surveillance. Apart from its well-established roles in controlling cell proliferation and cell death, numerous studies have proved that the Hippo pathway also functions as a key mechanotransducer to sense mechanical changes in the microenvironment. Despite the identification of multiple essential mechanosensitive signaling molecules including RAP2, MAP4K, Agrin, and Spectrin, it remains poorly understood how mechanical stimuli are transmitted from plasma membrane localized receptors to activate Hippo signaling cascade-mediated cellular responses, especially in intact tissues. This study, through a genetic screen in Drosophila, uncovered a regulatory mechanism whereby mechanical tension drives tumor-suppressive cell competition though the Hippo pathway. The genetic and biochemistry data uncovered Toll-6 as an essential regulator of Hippo signaling and further identified α-Spec as an essential downstream component that regulates cell competition via tension-mediated actomyosin activation. Moreover, this study further demonstrated that inter-organ communication is critical for the removal of precancerous cells at a systemic level and discovered fat body (FB)-derived Spz5 as a crucial ligand (Kong, 2022).

This study demonstrated that polarity-deficient oncogenic clones are eliminated through tension-dependent cell competition and has identified Toll-6 as a key membrane receptor that physically interacts and acts through α-Spec to activate the Hippo pathway. It has long been recognized that both extrinsic cues such as ligands and intrinsic factors such as stiffness and cell-cell contact-mediated mechanical cues can determine cell fate and affect cell proliferation, yet relatively little is known about how the cytoskeleton system contributes to the elimination of precancerous cells during cell competition in vivo. The data show that both &alpha-Spec and Rho1, two essential cytoskeleton regulators, accumulate and facilitate the elimination of scrib clones. In addition, α-Spec as a crucial linker that bridges Toll-6 activation-induced tensional changes to cytosolic Hippo pathway activation. Interestingly, studies in the mammalian system showed that RhoA (human Rho1 ortholog) is responsible for mechanical force-induced cell extrusion. Thus, a similar tension-mediated cell-elimination mechanism might exist in the mammalian system to actively remove unfitted precancerous cells (Kong, 2022).

TLRs play critical roles in the innate immune response. The Drosophila genome encodes nine TLRs, of which only Toll (Tl/Toll-1) has a clear function in innate immunity. Interestingly, a paradoxical role of Tl in regulating cell competition has been reported. Activation of Tl in polarity-deficient clones suppresses the elimination of losers, whereas in the Myc-induced cell competition model, increased Tl activity accelerates the elimination of losers. Apart from Tl, Toll-2, Toll-3, Toll-7, Toll-8, and Toll-9 have been implicated in regulating cell competition in different contexts, while Toll-4 and Toll-5 have little effect. It is noteworthy that none of above studies has investigated the role of Toll-6 in cell competition. The current data not only reveal Toll-6 as a crucial regulator of tumor-suppressive cell competition but also show how the mechanical tension-mediated Hippo cascade is initiated from the cell membrane through the Toll-6-&alpha-Spec axis. Notably, this study found that Toll-6 was not required for Myc-induced cell competition. Given that TLRs are highly conserved in vertebrates and the elimination of scrib-depleted cells also exists in the mammalian system, further experiments are necessary to determine whether analogous mechanisms exist in mammals and humans to regulate mechanical tension-induced, Hippo pathway-mediated tumor-suppressive cell competition (Kong, 2022).

Inter-organ communication is essential for proper development and homeostasis maintenance of multicellular organisms under both physiological and pathological conditions. The tumor progression process is also shaped by the interactions between tumor and other organs, including the immune system. Recent studies in Drosophila have provided insightful understanding of the complex crosstalk between organs during tumorigenesis. The FB is the major immune organ of Drosophila, and it has been shown that intestinal tumor progression or abdominal tumor transplantation promotes the wasting behavior of FBs. The current findings that the transcription of spz5 is increased in the FB from scrib clone bearing larvae to facilitate tumor-suppressive cell competition may provide an in vivo mechanistic understanding of the inter-organ communications between FBs and remotely colonized precancerous clones. Together, the ism) around the boundary between losers and winners, which recruits α-Spec and provokes Hippo pathway-dependent elimination of scrib-/- clones. Meanwhile, the presence of scrib-/- loser cells in the eye disc will trigger a systemic effect on the distal organs, including FBs, which results in the transcription upregulation and secretion of Spz5, in turn forming a feedforward loop to reinforce the tumor-suppressive cell competition by binding to Toll-6 (Kong, 2022).

Although biochemical and genetic data demonstrate that Toll-6 physically interacts with α-Spec and that α-Spec is required for the elimination of scrib clones, these experiments were unable to explain the molecular mechanisms by which Toll-6 recruits α-Spec and initiates the downstream signaling transduction. Another limitation is that because the substantial analysis relies heavily on genetics to infer mechanism, enough rigorous biochemistry data was not included to prove how the binding of Toll-6 with α-Spec triggers Hippo signaling activation. Additionally, this study found that FB-derived Spz5 is essential for the elimination of scrib clones through inter-organ communications, but it is not understood completely how the spz5 mRNA level is upregulated systematically in the FBs of larvae that bear scrib mutant clones. Future work will be required to dissect the transcriptome changes of FBs upon precancerous clone induction in distal organs. Finally, this study showed that Spz5 acts through Toll-6 to regulate cell competition, and it is known that Spz5 can bind to other TLRs to regulate both cell death and survival through a three-tier mechanism (Foldi, 2017), suggesting that Spz5 can trigger intracellular signal transduction through ligand receptor binding. Nonetheless, a question that remains unsolved is why a signaling network that relays cell mechanical properties (Toll-6-α-Spec axis) should be regulated by a chemical ligand/receptor interaction; it would be interesting to further explore the underlying mechanisms (Kong, 2022).

Mechanisms underlying the cooperation between loss of epithelial polarity and Notch signaling during neoplastic growth in Drosophila

Aggressive neoplastic growth can be initiated by a limited number of genetic alterations, such as the well-established cooperation between loss of cell architecture and hyperactive signaling pathways. However, understanding of how these different alterations interact and influence each other remains very incomplete. Using Drosophila paradigms of imaginal wing disc epithelial growth, this study monitored the changes in Notch pathway activity according to the polarity status of cells (scrib mutant). The scrib mutation was shown to impact the direct transcriptional output of the Notch pathway, without altering the global distribution of Su(H), the Notch dedicated transcription factor. The Notch-dependent neoplasms require however, the action of a group of transcription factors, similar to those previously identified for Ras/scrib neoplasm (namely AP-1, Stat92E, Ftz-F1, and bZIP factors), further suggesting the importance of this transcription factor network during neoplastic growth. Finally this work highlights some Notch/scrib specificities, in particular the role of the PAR domain containing bZIP transcription factor and Notch direct target Pdp1 for neoplastic growth (Logeay, 2022).

In this study, using Notch-driven paradigms of epithelial overgrowth in Drosophila wing discs, the molecular mechanisms are described underlying the cooperation between Notch and polarity loss during neoplasia. It was shown that epithelial polarity alterations redirect the transcriptional outcome of the Notch signaling pathway, thus defining a specific set of new neoplastic Notch direct targets. It was further shown that this redirection occurs mainly on pre-existing Su(H)-bound regions rather than new ones. Finally, it was shown that, similar to what was previously described for Ras signaling, the AP-1/Stat/Yki/Ftz-f1 transcription factors are required for the cooperation between Notch signaling and polarity loss during neoplastic growth (Logeay, 2022).

Although cancer genomes exhibit multiple mutations in cancer cells, their functional interactions remain difficult to monitor and model. Neoplastic tissues, generated upon the combination of Notch pathway activation and polarity loss through scrib mutation, experience many cellular stresses: DNA-damage responses, but also endoplasmic reticulum and unfolded protein response, starvation or oxidative stresses. However, even though present, these different stresses, in particular oxidative stress and DNA damage, are not individually necessary in the context of polarity loss as blocking them or the cellular response they promote (by CAT/SOD overexpression, or inhibition of p53) did not significantly suppress the NS tumorous behaviors. These observations suggest that the different stress pathways activated during polarity loss are either not required for fueling growth (they are rather a consequence than a cause of neoplastic growth), or might act 'redundantly' to activate a common core response required for increased growth (Logeay, 2022).

Although Drosophila and mouse models have demonstrated that overactive signaling pathways cooperate with epithelial polarity impairment to generate neoplastic growth, the vast majority of studies seeking to understand the underlying mechanisms have focused primarily on the cooperation between activated RasV12 and scrib mutants, especially in Drosophila. Importantly, the current study, investigating the cooperation between Notch and polarity, shows that many observations made for Ras can be extended to Notch, suggesting that the paradigms used are not specific to Ras but might represent a more general tumor growth paradigm. But, because the main, if not only, Notch pathway outcome is transcriptional, the NICD/scrib- model allowed the modes of cooperation to be studied in greater detail. The cooperation between Notch pathway activation and polarity loss led to a specific transcriptional program, and in particular the activation of new Notch direct targets. This was not the consequence of a general redeployment to new target gene loci of Su(H), the Notch pathway-dedicated transcription factor, ruling out one possible model for the oncogene/polarity cooperation. Thus, what could be the mechanisms controlling which genes were activated in the different conditions? All 'Notch'-activating transcriptional complexes comprise NICD, Mastermind and Su(H). Although no differences in overall levels of NICD and Su(H) could be detected by western blot (data not shown), they could be modified in different ways post-translationally leading to different Notch responses (e.g. core Notch response, N-only, NS-only). Indeed, recent reports point towards different post-translational modifications for Su(H). However, whether they lead to different transcriptional programs, and whether they occur in vivo in the N and NS models, remain to be studied. Through the use of iRegulon, we demonstrated that the genes of the NS transcriptome, and most importantly the NS Notch direct targets, were enriched in their regulatory regions for elements corresponding to specific transcription factors, and in particular Stat92E or bZIP factors. The fact that similar transcription factor families were found in the overall group of upregulated genes and in the more limited subset of Notch direct genes suggests that the Notch output was controlled, at least in part, by factors that act more broadly on the genome. These analyses support a model in which polarity loss redirects the output of the Notch transcriptional program by the action of cooperating transcription factors. However, further work, such as detailed comparative ChIP analyses of the different factors in the different conditions, is required to establish this model firmly (Logeay, 2022).

Although this study demonstrated the involvement of a similar 'oncogenic module' as identified for the RasV12/scrib- neoplastic model, there are specifics that are likely oncogene specific. First, unlike what was reported for RasV12/scrib- transcriptomes, Yki/Sd/TEAD modules were not found to be enriched in the different Notch and scrib- transcriptomes. In the case of Ras, it has been shown that Yki activity can reprogram Ras by promoting the expression of the Ras pathway-specific regulators Capicua and Pointed to promote aggressive growth. Both genes were either unaffected (capicua) or downregulated (pointed) in the NS Notch-driven neoplastic paradigm, suggesting that, even though Yki is clearly required, changes in the expression of capicua and pointed are unlikely to be mediators here. These differing results in the enrichment of Yki/Sd/TEAD motifs between Notch and Ras transcriptomes in the context of polarity loss might reflect the inhibitory effect Notch has on Yki activity in the wing pouch, in part through the action of vestigial. Furthermore, in the NS transcriptome, a contribution was identified of the E(spl) bHLH transcriptional repressors, canonical Notch targets, which represents thus a Notch specificity. However, the fact that motifs for E(spl)-HLH repressors are found in the upregulated transcriptome of NS and not N could suggest that in NS the repressive ability of E(spl)-HLH factors is antagonized, further allowing higher expression of Notch targets. More precisely, the previous work identified many incoherent feed-forward loops in the N hyperplastic transcriptome, including through the action of E(spl) repressors, which might thus be resolved in NS. It would be interesting to explore further the link between NS and E(spl)-HLH-mediated repression, but due to the high redundancy between the seven E(spl)-HLH factors (Δ, &gamma', β, 3, 5, 7, 8) and Dpn, the requirement of E(spl)-HLH-mediated repression in Notch-driven neoplasia could not be formally tested (Logeay, 2022).

By performing functional assays to identify the genes and processes required for NS tumor growth, this study demonstrated that the Notch direct targets associated with 'de novo' NS-specific Su(H) peaks were unlikely to be major contributors. We did show, however, that the bZIP PAR domain-containing factor Pdp1 is required for NS tumor growth and invasiveness. Su(H) is bound in the vicinity of Pdp1 in all wing discs set-ups, and in particular in N and NS, and Pdp1 represents a 'core' Notch target activated in all overgrowth conditions, albeit at higher levels in polarity-deficient conditions. Pdp1 is not only a Notch target, but also a Jak/Stat target, at least in the developing eye, and canonical tandem Stat92E putative binding sites are found in its second intron, although not overlapping with Su(H) binding, which is found in its first intron. Interestingly, Pdp1 is required for Stat92E phosphorylation and efficient Jak/Stat signaling, suggesting that Notch might amplify Stat92E signaling during wing disc neoplastic growth, both through ligand expression (Upd ligands are Notch direct targets) and Pdp1 expression (Logeay, 2022).

Although Pdp1 downregulation could suppress NS neoplastic growth, it was not as efficient as JNK inhibition, or Yki downregulation, suggesting that other factors in parallel to Pdp1 might be involved, such as the previously identified Atf3, but also the other Notch direct target Ets21C. Indeed, RNAi-mediated knockdown of Atf3 or Ets21C partly suppressed Bx>NS tumor growth (GFP) and invasiveness (Mmp1). This action of both Pdp1 and Ets21C suggest a feed-forward loop downstream of Notch that in the context of polarity loss and JNK activity promotes neoplastic growth. However, given that Atf3, Pdp1 and Ets21C (but also Ftz-f1) are all upregulated in N hyperplastic conditions, their sole upregulation cannot be sufficient for neoplasia. The fact that Atf3 and Pdp1 iRegulon enrichments are not found in N could indicate that, despite being upregulated in hyperplastic N, their transcriptional activities are hindered, or that one key cooperating factor enabling their action is missing. Further studies are thus required to test this possibility and study how, in the context of normal epithelial polarity, Notch activation prevents the action of Pdp1/Ets21C/Atf3, thus preventing the transition to neoplasia (Logeay, 2022).

Hyperinsulinemia drives epithelial tumorigenesis by abrogating cell competition

Metabolic diseases such as type 2 diabetes are associated with increased cancer incidence. This study shows that hyperinsulinemia promotes epithelial tumorigenesis by abrogating cell competition. In Drosophila eye imaginal epithelium, oncogenic scribble (scrib) mutant cells are eliminated by cell competition when surrounded by wild-type cells. Through a genetic screen, this study found that flies heterozygous for the insulin receptor substrate chico allow scrib cells to evade cell competition and develop into tumors. Intriguingly, chico is required in the brain's insulin-producing cells (IPCs) to execute cell competition remotely. Mechanistically, chico downregulation in IPCs causes hyperinsulinemia by upregulating a Drosophila insulin Dilp2, which activates insulin-mTOR signaling and thus boosts protein synthesis in scrib cells. A diet-induced increase in insulin levels also triggers scrib tumorigenesis, and pharmacological repression of protein synthesis prevents hyperinsulinemia-induced scrib overgrowth. These findings provide an in vivo mechanistic link between metabolic disease and cancer risk via systemic regulation of cell competition (Sanaki, 2020).

Metabolic diseases such as type 2 diabetes and obesity are often accompanied by hyperinsulinemia, which is characterized by high levels of circulating insulin. In epidemiology, hyperinsulinemia has been implicated in increased cancer incidence. For instance, the risk of liver, pancreas, endometrium, kidney, and bladder cancers increases 1.5- to 2-fold in people with hyperinsulinemia. Although previous studies in Drosophila and rodents unveiled some aspects of the mechanism by which hyperinsulinemia promotes tumor growth and malignancy, the underlying mechanisms are still largely unknown (Sanaki, 2020).

Most cancers originate from epithelial cells that frequently lose apicobasal polarity during tumor progression. In Drosophila imaginal epithelium, loss-of-function mutations in evolutionarily conserved apicobasal polarity genes, such as scrib or discs large (dlg), disrupt epithelial integrity and result in tumorous overgrowth. Intriguingly, such oncogenic polarity-deficient cells do not overproliferate but are eliminated from the tissue when surrounded by wild-type cells, a phenomenon called tumor-suppressive cell competition. Previous work found multiple mechanisms that drive this cell elimination via cell-cell interaction between scrib and wild-type cells, which include Sas-PTP10D ligand-receptor interaction, Slit-Robo2-Ena/VASP-mediated scrib cell extrusion, and engulfment of scrib cells by wild-type cells. Through a genetic screen in Drosophila, this study found an unexpected new regulatory mechanism whereby hyperinsulinemia systemically abrogates tumor-suppressive cell competition and thus causes tumorigenesis in the epithelium. These data could provide a mechanistic explanation for the epidemiological evidence that links hyperinsulinemia and cancer incidence, thus contributing to a better understanding of cancer biology in vivo (Sanaki, 2020).

This study found that hyperinsulinemia in flies systemically suppresses cell competition in the eye epithelium, leading to tumorous overproliferation of polarity-deficient cells that are normally eliminated when surrounded by wild-type cells. It has been reported that high-sugar diet promotes tumor growth and metastasis of fly tumors with elevated Ras and Src signaling, providing a model of how abnormal physiology promotes tumor progression. In addition, studies in mice have shown that high-fat diet-induced obesity suppresses extrusion of oncogenic RasV12-expressing cells from mice intestine and that endogenous hyperinsulinemia contributes to pancreatic ductal adenocarcinoma. Thus, abnormal physiology, especially hyperinsulinemia, has a promotive effect on tumor development and progression, yet the mechanism by which hyperinsulinemia controls the initial step of tumorigenesis has been unclear. The current observations indicate that chico heterozygous mutant or IPCs-specific chico-knockdown larvae can be used as a Drosophila model of hyperinsulinemia. Consistently, although chico homozygous mutant flies drastically decrease their body weight, chico heterozygous mutant flies show increased body weight, implying a phenotypic outcome of hyperinsulinemia (Sanaki, 2020).

The findings that hyperinsulinemia systemically abrogates tumor-suppressive cell competition by boosting InR-TOR-mediated protein synthesis in pre-malignant cells may provide an in vivo mechanistic link between metabolic diseases and cancer risk. Previous work has shown that Sas-PTP10D signaling in scrib cells promotes their elimination by repressing epidermal growth factor receptor (EGFR) signaling. Defects in Sas-PTP10D signaling attenuates scrib cell elimination via cooperation between EGFR-Ras and TNF-JNK signaling, which leads to activation of the Hippo pathway effector Yorkie (Yki). On the other hand, this study found that hyperinsulinemia attenuates scrib cell elimination by fueling insulin-mTor signaling. Given that these two signaling pathways are independent, there would be no direct cross talk between Sas-PTP10D signaling and hyperinsulinemia-driven tumorigenesis. Rather, it is possible that both Sas-PTP10D inactivation (Yki activation) and insulin signaling activation (Tor activation) lead to the same biological outcome, namely, elevation of protein synthesis, which could explain how insulin signaling overrides Sas-PTP10D signaling (Sanaki, 2020).

Notably, differential levels of protein synthesis between cells has long been implicated in regulating classical Minute cell competition, which is a competitive elimination of cells with a heterozygous mutation for a ribosomal protein gene. In addition, recent work has found that losers of cell competition triggered by different mutations such as Minute, Myc, Mahjong, and Hel25E commonly show lower protein synthesis levels than that neighboring winners do (Nagata, 2019). Moreover, insulin-TOR signaling has been shown to control cell competition during mouse embryonic development. These observations suggest that differential levels of insulin-TOR signaling and protein synthesis between cells are the key for cell competition. Supporting this notion, scrib-induced cell competition can be compromised either by introducing Minute mutation in wild-type winners or by overexpressing Myc in scrib losers. These data show that scrib cells are insensitive to environmental insulin and thus are lower in insulin-TOR signaling and protein synthesis levels compared with that of the neighbors, and hyperinsulinemia reverses this balance and causes scrib tumorigenesis. Given that a drug treatment targeting cellular metabolism could prevent hyperinsulinemia-driven tumorigenesis, cancer risk risen by metabolic diseases may become controllable in the future (Sanaki, 2020).

Dynamic MAPK signaling activity underlies a transition from growth arrest to proliferation in Drosophila scribble mutant tumors

Human tumors exhibit plasticity and evolving capacity over time. It is difficult to study the mechanisms of how tumors change over time in human patients, in particular during the early stages when a few oncogenic cells are barely detectable. This study used a Drosophila tumor model caused by loss of Scribble (Scrib), a highly conserved apicobasal cell polarity gene, to investigate the spatial-temporal dynamics of early tumorigenesis events. The fly scrib mutant tumors have been successfully used to model many aspects of tumorigenesis processes. However, it is still unknown whether the fly scrib mutant tumors exhibit plasticity and evolvability along the temporal axis. This study found that the scrib mutant tumors display different growth rates and cell cycle profiles over time, indicative of a growth arrest-to-proliferation transition as the scrib mutant tumors progress. Longitudinal bulk and single-cell transcriptomic analysis of the scrib mutant tumors revealed that the MAPK pathway, including the JNK and ERK signaling activities, shows quantitative changes over time. High JNK signaling activity causes G2/M cell cycle arrest in the early scrib mutant tumors. In addition, JNK signaling activity displays a radial polarity with the JNK(high) cells located at the periphery of the scrib mutant tumors, providing an inherent mechanism that leads to an overall JNK signaling activity decrease over time. The ERK signaling activity, in contrast to JNK activity, increases over time and promotes growth in the late-stage scrib mutant tumors. Finally, high JNK signaling activity represses ERK signaling activity in the early scrib mutant tumors. Together, these data demonstrated that dynamic MAPK signaling activity, fueled by intratumor heterogeneity derived from tissue topological differences, drives a growth arrest-to-proliferation transition in the scrib mutant tumors (Ji, 2019).

This study demonstrates that dynamic changes in JNK and ERK activities underlie a transition from a growth arrest state to a proliferation state over time in Drosophila scrib mutant tumors (Ji, 2019).

JNK signaling activation was found to exhibit a radial polarity, with JNKhigh cells located at the surface of homozygous scrib mutant tumors. The underlying reason for the heterogeneous JNK activation pattern is not yet known. One possible reason is that the active JNK ligand Eiger might be mostly provided from external sources. This might involve hemocytes or the fat body, as recently shown for another nTSG mutant alg3 . Another possible input is potential mechanical stresses that the tumor surface cells experience because JNK signaling is activated at leading-edge cells, which assemble supracellular actin cables and experience high actomyosin-dependent tensile force during dorsal closure (Ji, 2019).

JNK activation induced by injury in the wing imaginal discs also leads to cell cycle arrest at G2, which can be restored by overexpression of Cdc25/String. Expression of Cdc25/string(stg) and Cks30A anti-correlates with Mmp1 in the single-cell data. Moreover, growth arrest in early scrib mutant tumors could not be arrested by overexpression of String alone, indicating that the cell cycle defects in early scrib mutant tumors are probably mediated by multiple factors downstream of high JNK signaling activity (Ji, 2019).

Clonal scrib mutant cells are eliminated through cell competition when they are surrounded by wild-type neighbors. It is noteworthy that clonal scrib mutant cells are likely to be in a different state from growth-arrested homozygous scrib mutant cells. Studies have shown that overexpression of RasV12, NICD and p35 can effectively block the clonal scrib mutant cells from undergoing apoptosis induced by cell competition. This study found that overexpression of RasV12, NICD and p35 had little effect in relieving the early scrib mutant tumors from growth arrest. It is likely that interactions between clonal scrib mutant cells and wild-type cells during cell competition induce an additional layer of complexity into determination of clonal scrib mutant cell state, as the effects of JNK activation are known to be highly context dependent (Ji, 2019).

In human solid tumors, the tumor margin and core were also shown to experience different immune environments. This study highlights that tissue topological factors (peripheral versus center) can be an inherent source of diversity in cell populations in growing tumors and that dynamic signaling rewiring during the processes of early tumorigenesis does not necessarily require the generation of de novo mutations or new cell clones (Ji, 2019).

Scribble and Discs-large direct initial assembly and positioning of adherens junctions during the establishment of apical-basal polarity

Apical-basal polarity is a fundamental property of animal tissues. Drosophila embryos provide an outstanding model for defining mechanisms that initiate and maintain polarity. Polarity is initiated during cellularization, when cell-cell adherens junctions are positioned at the future boundary of apical and basolateral domains. Polarity maintenance then involves complementary and antagonistic interplay between apical and basal polarity complexes. The Scribble/Dlg module is well-known for promoting basolateral identity during polarity maintenance. This study reports a surprising role for Scribble/Dlg in polarity initiation, placing it near the top of the network-positioning adherens junctions. Scribble and Dlg are enriched in nascent adherens junctions, are essential for adherens junction positioning and supermolecular assembly, and also play a role in basal junction assembly. The hypotheses were tested for the underlying mechanisms, exploring potential effects on protein trafficking, cytoskeletal polarity or Par-1 localization/function. The data suggest that the Scribble/Dlg module plays multiple roles in polarity initiation. Different domains of Scribble contribute to these distinct roles. Together, these data reveal novel roles for Scribble/Dlg as master scaffolds regulating assembly of distinct junctional complexes at different times and places (Bonello, 2019).

Identifying the earliest symmetry-breaking events that initially position AJs, thereby setting the boundary between apical and basolateral domains, is a key aspect of understanding how polarity is established. This study reports that Scrib/Dlg, best known for roles as basolateral determinants during polarity maintenance, play a separate and surprising role in organizing AJs during polarity establishment, positioning them at the top of the polarity network (Bonello, 2019).

Scrib and Dlg are multidomain proteins with many partners, allowing them to serve diverse biological functions, from synaptogenesis to oriented cell division. The data reveal they play distinct roles during polarity establishment and polarity maintenance, likely engaging very different sets of binding partners. This is supported by the evolving localization pattern of Scrib/Dlg on the plasma membrane, with sequential co-localization with and roles in positioning AJ versus SJ proteins, suggesting the capacity to engage with and position distinct junctional and polarity proteins. These analyses also begin to dissect the underlying molecular basis. Scrib's PDZ domains are important for the precision of initial polarity establishment but are redundant with other mechanisms for polarity maintenance after gastrulation, though they regulate SJ positioning (Bonello, 2019).

AJs play a key role at the boundary between apical and basolateral domains, and building a functional junction is a multistep process. This includes assembling the core cadherin-catenin complex, positioning it, and supermolecular assembly. Assembly of the core complex appears to occur coincident with synthesis, and thus small puncta are already present before cellularization. As cellularization proceeds, these are captured at the apicolateral interface in a process requiring Baz, Cno, and an intact actin cytoskeleton, where they coalesce into SAJs, with ~1500 AJ complexes and 200 Baz proteins. Cadherin-catenin complexes form independently of either Baz or Cno, but AJ positioning and full supermolecular assembly depend on both. This study found that Scrib/Dlg are also key for AJ apicolateral retention and supermolecular assembly, although Arm and Cno remain associated in misplaced puncta, and thus core AJ complexes remain intact. Further, a second junctional complex that arise during polarity establishment, the BJs, also require Scrib/Dlg for its supermolecular organization. Unlike AJs, BJ organization is not dependent on other polarity determinants including Cno, Rap1 or Par-1. It will be of interest to examine if Scrib/Dlg act via known regulators of cadherin clustering, including intrinsic (e.g., cis- and trans-interactions of cadherins) and extrinsic (e.g., local actin regulation, endocytosis) factors (Bonello, 2019).

The ultimate goal is to define molecular mechanisms underlying polarity establishment. The new data place Scrib/Dlg in a critical position near the top of the network, but also suggest they act via multiple effectors. Perhaps the strongest evidence for multiple roles with distinct effectors comes from analysis of scrib4. Supermolecular organization of both SAJs and BJ must involve interactions with specific partners via the PDZ domains- one speculative possibility is that these include core AJ proteins, as βcatenin can coIP with Scribble and interact with PDZ domains 1 and 4. Testing this idea will be an important future direction. This initial role may also involve modulating Par-1. During cellularization, Scrib/Dlg and Par-1 localize in 'inverse gradients': Scrib and Dlg enriched at the SAJ level, and Par-1 with higher cortical intensity basolaterally. Scrib/Dlg play a role in effective membrane recruitment of Par-1 at this stage, and effects of par-1-RNAi on SAJ protein localization during cellularization are largely similar to those of scrib-RNAi. However, regulating Par-1 is not the only mechanism by which Scrib/Dlg act, as AJs are rescued during gastrulation after par-1-RNAi (Bonello, 2019).

Scrib then plays a second PDZ-independent role as gastrulation begins, ensuring focusing of cadherin-catenin complexes and Baz into apical belt AJs. This requires the N-terminal LRRs but not the PDZs. Positioning Baz at this stage involves at least two inputs which are redundant with one another, one via Par-1 and one via an apical transport mechanism. One speculative possibility is that Scrib/Dlg also regulate protein trafficking, a role they have in other contexts. However, disrupting Scrib/Dlg function has very different consequences than disrupting Rab5-dependent trafficking, suggesting they do not act via Rab5. aPKC also provides important cues at this stage-perhaps Scrib/Dlg regulate aPKC localization or function. It will be important to further explore the nature of this second role (Bonello, 2019).

The initial goal more than a decade ago was to define roles of AJs in polarity establishment. However, it rapidly became apparent AJs are not at the top of the hierarchy. Cno, Rap1 and Baz act upstream of AJ positioning and supermolecular assembly. The new data moves understanding another step upward in the network, revealing a key role for Scrib/Dlg in regulating AJ positioning and assembly. However, they also reveal that the process is not a simple linear pathway, and raise new questions. Loss of Scrib or Dlg almost completely disrupts AJs during cellularization. However, effects on Baz and Cno are less complete-supermolecular assembly is affected, but they are retained in the apical half of the membrane. This suggests other cues are involved. The ultimate polarizing cue during syncytial development is the oocyte membrane, which then directs cytoskeletal polarization. Cytoskeletal cues regulate Cno localization. While the data rule out a role for Scrib/Dlg in establishing basic cytoskeletal polarity, they do not rule out roles, for example, in localizing a special 'type' of actin cytoskeleton in the apical domain. Retention of Cno at the membrane after Scrib/Dlg knockdown suggests that minimally basal Rap1 activity remains intact. Changes to early Par-1, and to a lesser extent Baz, cortical localization with loss of Scrib/Dlg, also raise the possibility that lipid-based regulation is impaired. At this time, it is not known what cues regulate Scrib/Dlg apical enrichment but AJs do not appear to direct this, nor are they essential for polarizing Cno or Baz. Continued characterization of the full protein network and molecular mechanisms governing polarity establishment will keep the field busy for years to come (Bonello, 2019).

Epithelial cells release adenosine to promote local TNF production in response to polarity disruption

Disruption of epithelial integrity contributes to chronic inflammatory disorders through persistent activation of stress signalling. This study uncovered a mechanism whereby disruption of apico-basal polarity promotes stress signalling. Depletion of Scribbled (Scrib), a baso-lateral determinant, causes epithelial cells to release adenosine through equilibrative channels into the extracellular space. Autocrine activation of the adenosine receptor leads to transcriptional upregulation of TNF, which in turn boosts the activity of JNK signalling. Thus, disruption of cell polarity feeds into a well-established stress pathway through the intermediary of an adenosine signalling branch. Although this regulatory input could help ensuring an effective response to acute polarity stress, it is suggested that it becomes deleterious in situations of low-grade chronic disruption by provoking a private inflammatory-like TNF-driven response within the polarity-deficient epithelium (Poernbacher, 2018).

This study has shown that adenosine acts as a warning signal in response to sub-apoptotic perturbation of polarity in epithelial cells. Adenosine released in the extracellular space triggers the local production of TNF, which in turn activates JNK, a well-established stress mediator. Because extracellular adenosine (e-Ado) is short-lived, it is unlikely to act at a long range. In fact, it is suggested that a large number of contiguous cells need to be disrupted for them to collectively release adenosine at a level that is sufficient to elicit a response. This community effect would ensure that a stress response is only mounted when a significant amount of tissue is affected, as would occur in a situation of low-grade but widespread stress. In this model of perturbation of cell polarity, adenosine promotes TNF production within the affected epithelium itself, eliciting a private inflammatory-like response without the involvement of immune cells. Such a local, e-Ado-stimulated, response could be an important contributor to pathologies associated with chronic epithelial damage. The above scenario is in contrast with the orthodox view that it is the activated monocytes/macrophages that normally produce TNF and that this is inhibited by adenosine. It is speculated that the type of response elicited by tissue disruption depends on the severity of insult or type of pathology, a consideration that must be kept in mind while designing therapeutic strategies (Poernbacher, 2018).

BTB-zinc finger oncogenes are required for Ras and Notch-driven tumorigenesis in Drosophila

During tumorigenesis, pathways that promote the epithelial-to-mesenchymal transition (EMT) can both facilitate metastasis and endow tumor cells with cancer stem cell properties. To gain a greater understanding of how these properties are interlinked in cancers, Drosophila epithelial tumor models were used, that are driven by orthologues of human oncogenes (activated alleles of Ras and Notch) in cooperation with the loss of the cell polarity regulator, scribbled (scrib). Within these tumors, both invasive, mesenchymal-like cell morphology and continual tumor overgrowth, are dependent upon Jun N-terminal kinase (JNK) activity. To identify JNK-dependent changes within the tumors a comparative microarray analysis was used to define a JNK gene signature common to both Ras and Notch-driven tumors. Amongst the JNK-dependent changes was a significant enrichment for BTB-Zinc Finger (ZF) domain genes, including chronologically inappropriate morphogenesis (chinmo). chinmo was upregulated by JNK within the tumors, and overexpression of chinmo with either RasV12 or Nintra was sufficient to promote JNK-independent epithelial tumor formation in the eye/antennal disc, and, in cooperation with RasV12, promote tumor formation in the adult midgut epithelium. Chinmo primes cells for oncogene-mediated transformation through blocking differentiation in the eye disc, and promoting an escargot-expressing stem or enteroblast cell state in the adult midgut. BTB-ZF genes are also required for Ras and Notch-driven overgrowth of scrib mutant tissue, since, although loss of chinmo alone did not significantly impede tumor development, when loss of chinmo was combined with loss of a functionally related BTB-ZF gene, abrupt, tumor overgrowth was significantly reduced. abrupt is not a JNK-induced gene, however, Abrupt is present in JNK-positive tumor cells, consistent with a JNK-associated oncogenic role. As some mammalian BTB-ZF proteins are also highly oncogenic, this work suggests that EMT-promoting signals in human cancers could similarly utilize networks of these proteins to promote cancer stem cell states (Doggett, 2015).

This report has defined the transcriptional changes induced by JNK signaling within both scrib>RasACT and scrib>NACT tumors by carrying out comparative microarray expression arrays. This analysis that JNK exerts a profound effect upon the transcriptional profile of both Ras and Notch-driven tumor types. The expression of nearly 1000 genes was altered by the expression of bskDN in either Ras or Notch-driven tumors, and less than half of these changes were shared between the two tumor types, indicating that JNK signaling elicits unique tumorigenic expression profiles depending upon the cooperating oncogenic signal. Nevertheless, of the 399 JNK-regulated probe sets shared between Ras and Notch-driven tumors, it is hypothesized that these had the potential to provide key insights into JNK's oncogenic activity, and to prioritize these targets, it was considered that the expression of the critical oncogenic regulators would not just be altered by bskDN, but would be normalized to close to wild type levels. This subset of the 399 probe set was identified by comparing the expression profile of each genotype back to control tissue, thereby producing a more focussed JNK signature of 103 genes. Notably, this included previously characterized targets of JNK in the tumors, such as Mmp1,cherand Pax, thereby providing validation of the approach. Also amongst these candidates were 4 BTB-ZF genes; two of which were upregulated by JNK in the tumors (chinmo and fru), and two downregulated (br and ttk) (Doggett, 2015).

Focussing upon chinmo, chinmo overexpression was shown to be sufficient to prime epithelial cells for cooperation with RasACT in both the eye antennal disc and in the adult midgut epithelium, and that chinmo is required for cooperative RasACTor NACT-driven tumor overgrowth, although its function was only exposed when its knockdown was combined with knockdown of a functionally similar BTB-ZF transcription factor, abrupt. This family of proteins is highly oncogenic in Drosophila, since previous work has shown that ab overexpression can cooperate with loss of scrib to promote neoplastic overgrowth, and in these studies, it was also shown that overexpression of a fru isoform normally expressed in the eye disc is capable of promoting cooperation with RasACT and NACT in the eye-antennal disc, in a similar manner to chinmo overexpression. Thus, whether fru also plays a role in driving Ras or Notch-driven tumorigenesis warrants further investigation. Indeed, a deeper understanding of the oncogenic activity of these genes is likely to be highly relevant to human tumors, since of the over 40 human BTB-ZF family members, many are implicated in both haematopoietic and epithelial cancers, functioning as either oncogenes (eg., Bcl6, BTB7) or tumor suppressors (eg., PLZF, HIC1). Furthermore, over-expression of BTB7, can also cooperate with activated Ras in transforming primary cells, and its loss makes MEFs refractory to transformation by various key oncogenes such as Myc, H-rasV12 and T-Ag, suggesting that cooperating mechanisms between BTB-ZF proteins and additional oncogenic stimuli might be conserved (Doggett, 2015).

JNK signaling in Drosophila tumors is known to promote tumor overgrowth through both the STAT and Hippo pathways. Deregulation of the STAT pathway was evident in the arrays through the upregulation of Upd ligands by JNK in both Ras and Notch-driven tumors. In contrast, although cher was identified in the arrays as being upregulated in both tumor types and previous studies have shown that cher is partly required for the deregulation of the Hippo pathway in scrib>RasACT tumors, more direct evidence for Hippo pathway deregulation amongst the JNK signature genes was lacking. In part, this could be due to JNK regulating the pathway through post-transcriptional mechanisms involving direct phosphorylation of pathway components. However, the failure to identify known Hippo pathway target genes, and proliferation response genes in general, may simply highlight limitations in the sensitivity of the array assay and the cut-offs used for determining significance, despite its obvious success in correctly identifying many known JNK targets (Doggett, 2015).

Whether tumor overgrowth through STAT and Yki activity is somehow associated with a stem cell or progenitor-like state remains uncertain. Although imaginal discs exhibit developmental plasticity and regeneration potential, and JNK signaling is required for both of these stem-like properties, there is no positive evidence for the existence of a population of asymmetrically dividing stem cells within imaginal discs. Instead, symmetrical divisions of progenitor cells may be the means by which imaginal discs can rapidly generate enough cells to form the differentiated structures of the adult fly. To date, progenitor cells have only been characterized in the eye disc neuroepithelium. These cells have a pseudostratified columnar epithelial morphology and express the MEIS family transcription factor, Hth, which is downregulated as cells initiate differentiation and begin expressing Dac and Eya. Interestingly, they also require Yki for their proliferation, and can be induced to overproliferate in response to increased STAT activity. However, analysis of cell fate markers indicated that tumor overgrowth was not likley to be solely due to the overproliferation of these undifferentiated progenitor cells. Although scrib>RasACT/NACT tumors, were characterized by the failure to transition to Dac/Eya expression in the eye disc, blocking JNK in scrib > RasACT/NACT tumors did not restore tumor cell differentiation, despite overgrowth being curtailed, and Hth expression was not maintained in the tumors in a JNK-dependent manner. Nevertheless, a JNK-induced gene such as chinmo is likely to be associated with promoting a progenitor-like state, since it is a potential STAT target gene required for adult eye development that is expressed in eye disc progenitor cells in response to increased Upd activity and its overexpression alone is sufficient to block Dac/Eya expression. Furthermore, chinmo is also required for cyst stem cell maintenance in the Drosophila testis, and the current work has shown that chinmo overexpression promotes increased numbers of esgGFP expressing stem cells or enteroblasts in the adult midgut. As the BTB-ZF protein Ab is also highly oncogenic and expressed in the eye disc progenitor cells, it is hypothesize that the JNK-induced expression ofchinmo in scrib>RasACT/NACT tumors could cooperate with Ab to maintain a progenitor-like cell state in the eye disc, and that this is required for scrib->RasACT/NACT tumor overgrowth. However, although Ab was expressed in chinmo-expressing, JNK positive tumor cells, Ab does not appear to be a JNK-induced gene. What JNK-independent mechanisms control ab expression will therefore require further analysis (Doggett, 2015).

Interestingly, previous studies have observed that ab overexpression in eye disc clones upregulates chinmo expression and although the effect of chinmo expression upon ab is yet to be described, the data at least suggest that the control of their expression is interlinked in a yet to be defined manner (Doggett, 2015).

Consistent with Chinmo being important for scrib->RasACT/NACTv tumor overgrowth, chinmo overexpression itself is also highly oncogenic. Over-expression of chinmo with RasACT or NACT drives tumorigenesis in the eye-antennal disc, and also resulted in enlarged brain lobes, presumably due to the generation of overexpressing clones within the neuroepithelium of the optic lobes. In the adult midgut, the overexpression of chinmo with RasACT in the stem cell and its immediate progeny, the enteroblast, promoted massive tumor overgrowth, resulting in esgGFP expressing cells completely filling the lumen of the gut, and eventual host lethality. The luminal filling of esgGFP cells is reminiscent of the effects of RasACT expression in larval adult midgut progenitor cells. Together with the data linking Chinmo function to stem or progenitor cells, these data reinforce the idea that epithelial tumorigenesis can be primed by signals, such as chinmo over-expression, that promote a stem or progenitor cell state (Doggett, 2015).

The function of some Drosophila BTB-ZF proteins including Chinmo and Ab, has also been linked to heterochronic roles involving the conserved let-7 miRNA pathway and hormone signals, to regulate the timing of differentiation. Indeed, Ab can directly bind to the steroid hormone receptor co-activator Taiman (Tai or AIB1/SRC3 in humans), to represses the transcriptional response to ecdysone signaling. Thus, the capacity of BTB-ZF proteins to influence the timing of developmental transitions, particularly if they impede developmental transitions within stem or progenitor cells, could help account for their potent oncogenic activity. Indeed, ecdysone-response genes were repressed by JNK in the tumorigenic state, consistent with the failure of the larvae to pupate and a delay in developmental timing. Whether repressing the ecdysone response cell autonomously might contribute to tumor overgrowth and/or invasion will be an interesting area of future investigation, given the complex role of hormone signaling in mammalian stem cell biology and cancers (Doggett, 2015).

Previous studies have suggested that JNK-dependent tumor cell invasion is developmentally similar to the JNK-induced EMT-like events occurring during imaginal disc eversion. Thus the capacity of JNK to also promote tumor overgrowth is reminiscent of how EMT inducers such as Twist (Twi) and Snail (Sna) are associated with the acquisition of cancer stem cell properties. In Drosophila, however, twi and snawere not induced by JNK in the tumors, although transcription factors involved in mesoderm specification, including the NF-kappaB homologue, dl (a member of the 103 JNK signature), and Mef2 (a member of the 399 JNK signature), were amongst the up-regulated JNK targets. Mesoderm specification is not necessarily associated with a mesenchymal-like cell morphology, however, dl is involved in the induction of EMT during embryonic development, and both dl and Mef2 act with Twi and Sna to coordinate mesoderm formation. Interestingly, recent studies have identified dl in an overexpression screen for genes capable of cooperating with scrib > in Drosophila tumorigenesis, and Mef2 has been identified as a cooperating oncogene in Drosophila, and possibly also in humans, where a correlation exists between the expression of Notch and Mef2 paralogues in human breast tumor samples. It is therefore possible that dl and Mef2 either act in combination with Twi or Sna, or independently of them but in a similar oncogenic capacity, to promote a mesodermal cell fate in scrib > RasACT/NACT tumors. The potential relevance of this to the mesenchymal cell morphology associated with tumor cell invasion, as well as the acquisition of progenitor states is worthy of further investigation (Doggett, 2015).

In mef2-driven tumors both overgrowth and invasion depend upon activation of JNK signaling, suggesting that Mef2 is not capable of promoting invasive capabilities independent of JNK. In contrast, chinmo+RasACT/NACT tumors appeared non-invasive and retained epithelial morphology despite the massive overgrowth, although closer examination of cell polarity markers will be required to confirm this. Furthermore, the overgrowth of chinmo+RasACT/NACT tumors was not dependent upon JNK signaling, suggesting that the maintenance of a progenitor-like state could be uncoupled from JNK-induced EMT-effectors associated with invasion. Whether clear divisions between mesenchymal behaviour and progenitor states in tumors can be clearly separated in this manner is not yet clear, however, overall, it is likely that multiple JNK-regulated genes will participate in both promoting tumor overgrowth as well as migration/invasion. Although this study used the 103 JNK signature as a means to focus upon potential key candidates, an analysis of the 399 JNK-regulated probe sets common to both Ras and Notch-driven tumours has the potential to provide deeper insights into the multiple effectors of JNK signaling during tumorigenesis. Whilst the individual role of these genes can be probed with knockdowns, the complexity of the response, potentially with multiple redundancies and cross-talk, will ultimately need a network level of understanding to more fully expose key nodes participating in overgrowth and invasion. This approach has considerable potential to further expose core principles and mechanisms that drive human tumorigenesis, since it is clear that many fundamental commonalities underlie the development of tumors in Drosophila and mammals (Doggett, 2015).

The neuronal protein Neurexin directly interacts with the Scribble-Pix complex to stimulate F-actin assembly for synaptic vesicle clustering

Synaptic vesicles (SVs) form distinct pools at synaptic terminals, and this well-regulated separation is necessary for normal neuro-transmission. However, how SV cluster in particular synaptic compartments to maintain normal neurotransmitter release remains a mystery. The presynaptic protein Neurexin (NRX) plays a significant role in synaptic architecture and function, and some evidences suggest that NRX is associated with neurological disorders, including autism spectrum disorders. However, the role of NRX in SV clustering is unclear. Using the neuromuscular junction at the 2-3 instar stages of Drosophila larvae as a model and biochemical, imaging, and electrophysiology techniques, this study demonstrate that Drosophila NRX (DNRX) plays critical roles in regulating synaptic terminal clustering and release of SVs. DNRX controls the terminal clustering and release of SVs by stimulating presynaptic F-actin. Furthermore, the results indicate that DNRX functions through the scaffold protein Scribble and the GEF protein Pixie to activate the small GTPase Rac1. A direct interaction was observed between the C-terminal PDZ-binding motif of DNRX and the PDZ domains of Scribble and that Scribble bridges DNRX to DPix, forming a DNRX/Scribble/DPix complex that activates Rac1 and subsequently stimulates presynaptic F-actin assembly and SV clustering. Taken together, this work provides important insights into the function of DNRX in regulating SV clustering, which could help inform further research into pathological neurexin-mediated mechanisms in neurological disorders such as autism (Rui, 2017).

Neurons are the basic unit of the nervous system, and they communicate with each other through synapses. After being captured by sensory organs, neural signals pass between synapses in the form of neurotransmitters. As the vehicles of neurotransmitters, synaptic vesicles (SV) are essential for neurotransmission. SVs can be divided into three distinct pools according to their localization and function. The SVs adjacent to the active zone and ready to be released are referred to as the ready release pool. The second pool of vesicles is the exo/endo-cycling pool, which is also found close to release sites and supplies the ready release pool. Finally, the pool located away from the active zone, and that contains the majority of SVs, is referred to as the reserve pool and is considered to be a storage pool. Actin is a major part of the cytoskeleton that is required for maintaining the architecture of synapses as well as contributing to their function, and a significant amount of evidence has shown that the localization, translocation, and release of SVs can be altered by disturbing the polymerization of pre-synaptic actin (Rui, 2017).

The synapse is a highly specialized structure, and synaptogenesis is a highly complex process. Previous studies have shown that synaptic adhesive molecules play important roles in synaptogenesis and neurotransmission, and a number of synaptic cell adhesion molecules, including Neuroligins and Neurexins have been identified over the past few decades. Neurexin was first recognized as a receptor for α-latrotoxin, a black widow spider venom component that triggers massive neurotransmitter release. There are three neurexin genes in mammals, each of which has two promoters generating α-Neurexin and β-Neurexins, whereas there is only one neurexin-1 gene in Drosophila (dnrx). Recent studies both in Drosophila and mammals showed that Neurexin plays a significant role in synaptic architecture and function, and there is evidence suggesting that neurexin is associated with autism spectrum disorders (ASDs). The complexity and redundancy of the neurexin genes in mammals motivated the authors to focus on simpler model systems, such as Drosophila, to investigate the in vivo function of DNRX (Rui, 2017).

Neurexin has been shown to bind to several molecules, including the presynaptic scaffolding proteins Mint (Biederer, 2000), CASK (Sun, 2009), and LRRTM2 (de wit, 2009; Ko, 2009). Recently, DNRX also has been demonstrated to interact with the N-ethylmaleimide sensitive factor to regulate short-term synaptic depression and to interact with Spinophilin to maintain active zone architecture (Muhammad, 2015; Li, 2015). A typical trans-synaptic complex is formed by the heterophilic interaction of presynaptic Neurexins and postsynaptic Neuroligins, and these complexes have attracted much attention as scaffolding complexes that not only maintain the normal structure of the synapse but also function in passing signals across the synapses. It is thus clear that Neurexin is a multifunctional molecule. Despite the identification and characterization of these proteins that functionally associated with DNRX, understanding of the pathways including DNRX that control synaptic function are still incomplete, with other partners and mechanisms yet to be uncovered and analyzed (Rui, 2017).

This study has investigated the role of DNRX in the cluster and release of SVs at synaptic terminals. The effect of DNRX is mediated by presynaptic F-actin and there is a direct interaction between the C-terminal PDZ-binding motif of DNRX and the PDZ domains of the tumor suppressor protein Scribble. Furthermore, Scribble bridges DNRX to DPix, forming a DNRX-Scribble-DPix complex to activate Rac1 and affect presynaptic F-actin assembly and SV clustering. Taken together, these studies provide novel insight into the mechanisms underlying the regulation of neurotransmitter release by DNRX (Rui, 2017).

Neurexin is a highly conserved cell adhesion molecule that is predominantly localized at the presynaptic terminal. Previous studies have shown that Neurexin plays a significant role in synaptic architecture and function. In addition, accumulating evidence has implicated Neurexin in Autism Spectrum Disorders (ASDs). ASDs are neurodevelopmental disorders characterized by deficits in communication and social interaction as well as restricted interests and repetitive and stereotypic patterns of behavior. However, the precise function and underlying molecular mechanisms of Neurexin in both normal physiology and ASDs remain unclear. Drosophila Scribble is a cytoplasmic scaffolding protein that was first recognized as a tumor suppressor that regulates epithelial cell adhesion and migration in mammals. Recently, it has been shown that Scribble is also localized in the nervous system both in invertebrate and vertebrate animals, and plays a role in synaptic plasticity and animal behavior, including learning, memory, social behavior, and olfactory behavior. However, how Scribble functions at the synapse remains unknown. In this study, this study revealed that DNRX interacts directly with the Scribble PDZ domains through the very C-terminal PDZ-binding motif to regulate presynaptic F-actin and SVs (Rui, 2017).

F-actin is highly enriched at synaptic terminals and is vital for SV traffic, localization, and release For decades, F-actin emerged as the major cytoskeleton identified in presynaptic nerve terminals. Considering the especial location of F-actin at the internal space of presynaptic nerve terminals, it raised a hypothesis that F-actin regulates synaptic vesicle localization and release. Previous studies from several groups provided consistent evidence that after disrupting the polymerization of actin, pre-synapse affected SV traffic, localization, and neurotransmitter release. Moreover, cortical actin has been identified as a barrier for vesicles during the process of moving to the active zone in the presynaptic terminal. However, whether and how F-actin participates in the regulation of SV at synapse is still controversial. Therefore, further investigation will be necessary to determine the function of F-actin at synapse and especially for advances in understanding of the relationship with SV. By now the function of F-actin at synapse is still poorly understood. To better understand the effect of F-actin on SV this study assessed the cluster and release of SV after ablating the actin-associated genes and found the obvious defects of SV cluster and release. To further test this possibility, additional experiments are needed to elaborate the vital role of F-actin in SV regulation in future (Rui, 2017).

SV distribution and dynamics are essential for normal neural signal transmission and for synaptic plasticity both at peripheral and central synapses. Disruptions in SV function will lead to various forms of neurological disorders. The process of synaptic vesicle priming, docking, and fusion with the presynaptic membrane has been investigated extensively, and numerous molecules involved in this process have been identified. These include synaptotagmins, synapsins, synaptobrevins, and Munc18. However, how SV cluster in particular compartments and their role in regulating neurotransmitter release at the presynaptic terminal are unclear. The results from this study provide compelling evidences that DNRX plays an essential role in the distribution and release of SVs (Rui, 2017).

In this study, the data suggest the amount of F-actin is significantly reduced. Moreover, presynaptic Cortactin or the active form of DPak, key regulators of the actin cytoskeleton, are able to rescue the defects in SV distribution and spontaneous release frequency in the dnrx mutant. These results illustrate the essential role of presynaptic actin in regulating SVs localization and release. The results are consistent with the recent findings showing that Arp2/3 complex-mediated actin regulation is important for presynaptic neurotransmitter release (Rui, 2017).

Previous work has shown that Scribble is a scaffolding, tumor suppressor protein that through its PDZ domains interacts with a number of proteins, including β-catenin, βPix, and NOS1AP. Although NOS1AP can bind directly to the fourth Scribble PDZ domain, βPix has binding affinity to all four PDZ domains of Scribble. The present study used co-immunoprecipitation to show that DNRX can form a complex with Scribble in the Drosophila nervous system in vivo. DNRX and Scribble are co-localized in both central and peripheral nervous system during embryonic, larval, and adult stages. Importantly, these two proteins are highly expressed in the mushroom body of Drosophila, suggesting a key role in learning and memory. DNRX can directly bind to all four Scribble PDZ domains through its C-terminal PDZ-binding motif, similar to what is observed for βPix. These interaction results suggest that there may be competitions and cross-effect between DNRX and βPix. These interactions may also relate to the mutual effect on the protein level of Scribble and DNRX. Because the mRNA level of Scribble is not altered in the dnrx mutant, it is possible that when the DNRX or Scribble was absent, the complex becomes destabilized and subsequently degraded. Further experiments are needed to address this possibility (Rui, 2017).

What is the functional consequence of the Scribble and DNRX interaction? The present study provides evidence that Scribble may act as a bridge between DNRX and DPix to regulate the actin cytoskeleton. βPix is a GEF specific to Rac1/Cdc2, a key mediator of actin reorganization in response to various stimuli. In the mammalian system, Rac1 is locally activated in dendritic spines, and this spatial restricted activation is regulated by Pix (Zhang, 2005). The present study shows that the active form of Rac1 (Rac1-GTP) is reduced in the dnrx mutant, dnrx, and scribble knockdown flies compared with wild-type for the protein level of DPix in these lines were decreased, supporting the idea that the DNRX and Scribble interaction activates Rac1. Recent studies show that Rac1 plays a critical roles in animal behavior, particularly in the process of forgetting (36). In addition, Scribble has recently been reported to activate forgetting through Rac1 in Drosophila (35). Taken together, it is suggested that DNRX may also be involved in forgetting by regulating the Rac1 signaling pathway. Indeed, it has been demonstrated that Rac1 activation is defective in multiple autism-related gene mutations, including the dnrx gene, and that Rac1 has been proposed to be a converging node linked to ASD (36). Thus, the present study demonstrating that DNRX interacts with Scribble and DPix to regulate Rac1 provides direct mechanistic insight into not only the fundamental mechanisms underlying the roles of the neuroligin-neurexin complex in actin-mediated presynaptic regulation, but also the pathological mechanism of ASD (Rui, 2017).

Serpin facilitates tumor-suppressive cell competition by blocking Toll-mediated Yki activation in Drosophila

Normal epithelial tissue exerts an intrinsic tumor-suppressive effect against oncogenically transformed cells. In Drosophila imaginal epithelium, clones of oncogenic polarity-deficient cells mutant for scribble (scrib) or discs large (dlg) are eliminated by cell competition when surrounded by wild-type cells. In this study, a genetic screen in Drosophila identified Serpin5 (Spn5), a secreted negative regulator of Toll signaling, as a crucial factor for epithelial cells to eliminate scrib mutant clones from epithelium. Downregulation of Spn5 in wild-type cells leads to elevation of Toll signaling in neighboring scrib cells. Strikingly, forced activation of Toll signaling or Toll-related receptor (TRR) signaling in scrib clones transforms scrib cells from losers to supercompetitors, resulting in tumorous overgrowth of mutant clones. Mechanistically, Toll activation in scrib clones leads to c-Jun N-terminal kinase (JNK) activation and F-actin accumulation, which cause strong activation of the Hippo pathway effector Yorkie that blocks cell death and promotes cell proliferation. These data suggest that Spn5 secreted from normal epithelial cells acts as a component of the extracellular surveillance system that facilitates elimination of pre-malignant cells from epithelium (Katsukawa, 2018).

Clones of oncogenic polarity-deficient cells are actively eliminated from Drosophila imaginal epithelium when surrounded by normal tissue, indicating the existence of intrinsic tumor-suppression mechanism by cell competition. The present study shows that normal epithelial cells secrete Spn5 to facilitate the tumor-suppressive cell competition by antagonizing Toll signaling activation in polarity-deficient cells. Elevation of Toll signaling in polarity-deficient cells transforms them from losers to supercompetitors, which leads to tumorous overgrowth of mutant tissue. Thus, Spn5 acts as a component of the extracellular surveillance system that eliminates oncogenic cells by cell competition. It is not known at this stage why scrib cells are more sensitive to loss of spn5 to upregulate Toll signaling compared to surrounding wild-type cells. One possible mechanism that drives Toll activation in scrib cells would be JNK activation, which was shown to be sufficient to activate Toll signaling (Katsukawa, 2018).

Interestingly, it has been shown that activation of TRR signaling in losers of Myc- or Minute-induced cell competition causes losers' death through nuclear factor κB (NF-κB)-mediated induction of cell death gene hid or rpr, respectively. Consistent with this report, it has been shown in Drosophila larval fat bodies that activation of Toll signaling leads to inactivation of Yki, which may cause hid- or rpr-mediated cell death because one of the important Yki targets is a caspase inhibitor diap1. These observations intriguingly indicate that Toll signaling has opposite roles in different types of cell competition; while Toll activation promotes elimination of losers in Myc- or Minute-induced cell competition, it suppresses elimination of polarity-deficient losers in tumor-suppressive cell competition. Importantly, however, in both cases, Toll or TRR signaling seems to act as an oncogenic signaling that promotes expansion of pre-malignant winner clones within the tissue. Consistent with these findings in Drosophila, it has been reported that upregulation of Toll-like receptors (TLRs) is associated with tumor growth and progression in some human cancers. In addition, one of the human orthologs of Drosophila Spn5, SpnA5, has been shown to inhibit breast cancer growth and metastasis, and its expression level is decreased in renal cell carcinoma and sarcoma. These observations, together with the data from Drosophila genetics, suggest that Toll signaling drives tumorigenesis by promoting supercompetition of oncogenic cell clones (Katsukawa, 2018).

The mechanism by which Toll activation in polarity-deficient cells leads to Yki activation is an important open question for future studies. One possible mechanism is co-activation of JNK and Ras signaling in Toll-activated scrib cells, as these two pathways have shown to cooperate to induce Yki activation through F-actin accumulation and Wts inactivation. Interestingly, it has been shown in mammalian systems that the TLR signaling activates JNK signaling and that several TLRs activate EGFR-Ras signaling upon immune response. Given that signaling molecules identified in Drosophila are all conserved, similar Toll-mediated regulation of tumorigenesis could be involved in human cancer (Katsukawa, 2018).

Distinct activities of Scrib module proteins organize epithelial polarity

A polarized architecture is central to both epithelial structure and function. In many cells, polarity involves mutual antagonism between the Par complex and the Scribble (Scrib) module. While molecular mechanisms underlying Par-mediated apical determination are well-understood, how Scrib module proteins specify the basolateral domain remains unknown. This study demonstrates dependent and independent activities of Scrib, Discs-large (Dlg), and Lethal giant larvae (Lgl) using the Drosophila follicle epithelium. The data support a linear hierarchy for localization, but rule out previously proposed protein-protein interactions as essential for polarization. Cortical recruitment of Scrib does not require palmitoylation or polar phospholipid binding but instead an independent cortically stabilizing activity of Dlg. Scrib and Dlg do not directly antagonize atypical protein kinase C (aPKC), but may instead restrict aPKC localization by enabling the aPKC-inhibiting activity of Lgl. Importantly, while Scrib, Dlg, and Lgl are each required, all three together are not sufficient to antagonize the Par complex. These data demonstrate previously unappreciated diversity of function within the Scrib module and begin to define the elusive molecular functions of Scrib and Dlg (Khoury, 2020).

Despite being central regulators of cell polarity in numerous tissues from nematodes to mammals, the mechanisms of Scrib module activity have remained obscure. The current work highlights previously unappreciated specificity in these activities, and begins to define the molecular functions of Scrib, Dlg, and Lgl. The data focus on the Drosophila follicle epithelium, as well as in some cases Drosophila embryos, but it is important to note that tissue contexts can differ in polarity programs: For example, in the adult Drosophila midgut epithelium, where Scrib module proteins are dispensable for epithelial organization. The failure to detect phenotypic enhancement in double-mutant follicle cells, compared to single mutants, which together with the complete penetrance of single-mutant phenotypes suggest full codependence of function rather than functional overlap. Moreover, Scrib module mutants could not be bypassed in any combination by overexpression of other genes in the module, consistent with unique roles for each protein. Thus, while Scrib, Dlg, and Lgl act in a common 'basolateral polarity' pathway, they each contribute distinct functions to give rise to the basolateral domain (Khoury, 2020).

Cell polarity is particularly evident at the plasma membrane, and most polarity regulators act at the cell cortex. Therefore, a key question in the field has concerned the mechanisms that allow cortical localization of the Scrib module and Par complex proteins, which exhibit no classic membrane-association domains. A simple linear hierarchy was found for cortical localization in the follicle that places Dlg most upstream, and contrasts with that recently described in the adult midgut, where Scrib appears to be most upstream. This work highlights the requirement of Dlg for Scrib localization, and provides insight into the mechanism, in part by ruling out previous models. One model involves a direct physical interaction, mediated by the Scrib PDZ domains and Dlg GUK domain. However, in vivo analyses show that follicle cells mutant for alleles lacking either of these domains have normal polarity; these results are supported by data from imaginal discs. In contrast, this study showa that the SH3 domain is critical for Scrib cortical localization as well as polarity. The Dlg SH3 and GUK domains engage in an intramolecular 'autoinhibitory' interaction that negatively regulates binding of partners, such as Gukh and CASK. The dispensability of the GUK domain provides evidence against an essential role for this mode of regulation in epithelial polarity, and highlights the value of investigating the GUK-independent function of the Dlg SH3 (Khoury, 2020).

A second mechanism of Scrib cortical association was also excluded. Mammalian Scrib is S-palmitoylated and this modification is required for both cortical localization and function. As Drosophila Scrib was also recently shown to be palmitoylated, an appealing model would involve Dlg regulating this posttranslational modification. However, no changes to Scrib palmitoylation were detected in a dlg mutant, and chemically or genetically inhibiting Drosophila palmitoyltransferases also had no effect on Scrib localization, although the possibility that Scrib palmitoylation may be part of a multipart localization mechanism cannot be excluded. Surprisingly, palmitoylated Scrib is incapable of reaching the cortex in dlg mutants. While a constitutively myristoylated Scrib can bypass this requirement for localization, it is nevertheless insufficient to support polarity in the absence of Dlg. These results indicate that Dlg regulates additional basolateral activities beyond localizing Scrib (Khoury, 2020).

Lgl's role as an aPKC inhibitor is well-characterized, but how Scrib and Dlg influence this antagonism is not understood. This study shows that Scrib and Dlg maintain cortical Lgl by regulating its phosphorylation by aPKC, rather than by direct physical recruitment to the membrane. A contemporaneous study by Ventura (2020) supports this finding, further showing that the major factor in Lgl cortical stabilization is PIP2. The current data also suggest that the basolateral-promoting activities of Scrib and Dlg are not via direct inhibition of aPKC kinase activity or intrinsic antagonism of aPKC localization. Instead, they are consistent with models in which Scrib and Dlg regulate the three specific aPKC-targeted residues in Lgl. Previous work has demonstrated that these phosphorylated serines (656, 660, 664) are neither functionally nor kinetically equivalent, and a recent model proposes that S664 is required for basolateral polarization by mediating a phosphorylation-dependent interaction with the Dlg GUK domain. Beyond the dispensability of the GUK domain, the enhanced ability of LglAAS to inhibit aPKC and its ability to do so largely independently of Scrib and Dlg, argues against this model. Moreover, only LglAAS among the phospho-mutants can dominantly affect aPKC activity, while WT Lgl can do the same only if Scrib and Dlg are present. Together, these results suggest that S656 is the critical inhibitory residue whose phosphorylation must be limited to enable Lgl's activity (Khoury, 2020).

The mechanism by which LglS656A,LglS660A(AAS) (LglAAS) can suppress even constitutively active aPKCΔN remains unclear. aPKC substrates can act as competitive inhibitors; either an increased substrate affinity for aPKC or reduced ability to be inhibited by virtue of having fewer phosphorylation sites could make LglAAS a more effective inhibitor than WT Lgl. Supporting this idea, it was previously shown that S664, the only residue still available in LglAAS, is phosphorylated with higher kinetic preference than S656 or S660. It is also possible that some LglAAS phenotypes may be due to aPKC-independent effects resulting from reduced phosphorylation on S656 and S660. Nevertheless, a model is proposed in which Scrib and Dlg 'protect' Lgl by limiting phosphorylation of S656, thus tipping the inhibitory balance to allow Lgl to inhibit aPKC and establish the basolateral domain (Khoury, 2020).

What mechanism could underlie Scrib and Dlg protection of Lgl? One mechanism could involve generating a high phospholipid charge density at the basolateral membrane, which has been shown to desensitize Lgl to aPKC phosphorylation in vitro. However, the current data do not find evidence for regulation of phosphoinositides by Scrib and Dlg. A second possibility is that Scrib and Dlg could scaffold an additional factor, such as protein phosphatase 1, which counteracts aPKC phosphorylation of Lgl. Alternative mechanisms include those suggested by recent work on PAR-1 and PAR-2 in Caenorhabditis elegans zygotes, a circuit with several parallels to the Scrib module. In this system, PAR-2 protects PAR-1 at the cortex by shielding it from aPKC phosphorylation through physical interaction-dependent and -independent mechanisms. By analogy, binding with Scrib or Dlg could allosterically regulate Lgl to prevent phosphorylation, although this study has ruled out the Lgl-Dlg interaction documented in the literature. Scrib or Dlg might also act as a 'decoy substrate' for aPKC, as PAR-2 does in PAR-1 protection. Indeed, Scrib is phosphorylated on at least 13 residues in Drosophila embryos, although the functional relevance of this is not yet known (Khoury, 2020).

Overall, this work highlights the multifaceted nature of Scrib module function. The failure to bypass Scrib module mutants by transgenic supply of any single or double combination of other module components, including several that were constitutively membrane-tethered, suggests that every member contributes a specific activity to polarity. Nevertheless, even the simultaneous ectopic localization of all three Scrib module proteins was insufficient to disrupt the apical domain. This insufficiency in basolateral specification may reflect an inability of apical Scrib and Dlg to protect Lgl from aPKC phosphorylation, perhaps due to the distinct molecular composition of the apical and basolateral domains. This supports the idea that in addition to intrinsic activity via Lgl, the Scrib module must recruit or activate additional, as yet unidentified effectors in basolateral polarity establishment. The independent as well as cooperative activities of the Scrib module delineated in this study demonstrate previously unappreciated complexity in the determination of basolateral polarity and set the stage for future mechanistic studies of Scrib module function (Khoury, 2020).


REGULATION

Protein Interactions

Membrane-associated guanylate kinases (MAGUKs), such as Discs-large (Dlg), play critical roles in synapse maturation by regulating the assembly of synaptic multiprotein complexes. Previous studies have revealed a genetic interaction between Dlg and another PDZ scaffolding protein, Scribble (Scrib), during the establishment of cell polarity in developing epithelia. The biochemical nature of this interaction has remained elusive, raising questions regarding the mechanisms by which the actions of both proteins are coordinated. This study reports a new Dlg-interacting protein, GUK-holder (GUKh), that interacts with the GUK domain of Dlg and that is dynamically expressed during synaptic bouton budding. At Drosophila synapses Dlg colocalizes with Scrib and this colocalization is likely to be mediated by direct interactions between GUKh and the PDZ2 domain of Scrib. Dlg, GUKh, and Scrib form a tripartite complex at synapses, in which Dlg and GUKh are required for the proper synaptic localization of Scrib. These results provide a mechanism by which developmentally important PDZ-mediated complexes are associated at the synapse (Mathew, 2002).

In Drosophila, dlg mutants in which the GUK domain is absent exhibit abnormalities in synapse structure. Moreover, transgenic Dlg lacking the GUK domain fails to localize at synapses when expressed in a dlg mutant background. These findings imply that the GUK domain is required for a synaptic function and targeting of Dlg. To gain further insight on how the GUK domain of DLG exerts its various functions, proteins interacting with this domain were sought. GUK-holder, a novel synaptic protein contains a WH1/EVH1-like domain in its N-terminal half and a PDZ binding motif at its C terminus; the PDZ binding motiif has been identified as a GUK interactor. GUKh is expressed in a dynamic fashion during synaptic bouton formation. In addition, it also binds to a PDZ domain of Scribble (Scrib; a tumor suppressor protein interacts genetically with Dlg in developing epithelia) thus physically linking Dlg to Scrib. Indeed, coimmunoprecipitation analyses together with immunocytochemical studies on wild-type and mutant larvae provide strong evidence that Dlg, GUKh, and Scrib exist in a tripartite complex at the NMJ. Most notably, normal GUKh function is required for the synaptic localization of Scrib (Mathew, 2002).

Together, these yeast two-hybrid, coimmunoprecipitation, and colocalization studies provide compelling evidence that GUKh interacts with Dlg in vivo. This interaction is mediated by a region near the C terminus of GUKh. However, as revealed by genetic analysis, the synaptic localization of GUKh does not depend on Dlg. This suggests that domains other than the Dlg interacting motif may mediate its synaptic localization. For instance, the single WH1-like domain of GUKh might interact directly or indirectly with the synaptic cytoskeleton. WH1 domains in other proteins bind F-actin, actin-associated proteins such as zyxin, vinculin, and profilin, or the spectrin-bound scaffolding protein Shank/ProSAP. Association of GUKh with cytoskeletal elements might also be mediated by those sequences that exhibit moderate similarity to the actin binding protein Kelch (Mathew, 2002).

While an association of GUKh with the actin-based synaptic cytoskeleton currently remains hypothetical, the C-terminal tETAL motif specifically binds to the second PDZ domain of Scrib. Anatomical and biochemical experiments suggest that in vivo, Dlg, Scrib, and GUKh may exist in the same complex at the NMJ. Alternatively, the three proteins could interact pairwise, forming separate heterodimers. Since GUKh was found to still localize normally at dlg mutant NMJs, it is proposed that Dlg and GUKh act in concert rather than in a hierarchical manner to recruit Scrib. As a possible mechanism, binding to the GUK domain of Dlg could cause sterical changes in GUKh, such that the tETAL motif becomes available for interaction with Scrib. A caveat to this study is that hypomorphic gukh mutants were used, and therefore, a requirement of GUKh in Dlg localization cannot be ruled out (Mathew, 2002).

While Dlg and Scrib are colocalized along the rims of synaptic boutons, which, as has been demonstrated for Dlg, comprise both the presynaptic membrane and the postsynaptic junctional region (SSR), GUKh intersects that region only in a narrow strip. Yet, in budding boutons, GUKh displays a complementary pattern to Dlg. These observations suggest that GUKh may not be continuously bound to Dlg but rather may be involved in transient interactions. The process of bouton budding is a dynamic process that is characterized by equally dynamic changes in both GUKh and Dlg distribution. The accumulation of GUKh at the core of budding boutons and the disappearance of Dlg at the border of buds suggest that both proteins serve different roles during this process. Interestingly, FasII, a molecule that mediates synapse stabilization but that also imposes an adhesive constraint on synaptic growth, faithfully resembles the changes in distribution of Dlg during budding, consistent with a role for Dlg in synaptic localization. The presence of GUKh at budding regions may represent a role for this protein in destabilizing regions of the synaptic bouton, thereby allowing for bud formation (Mathew, 2002).

In contrast to GUKh, Scrib is expressed throughout the SSR in exact colocalization with Dlg. Nonetheless, Scrib localization at distal regions of the SSR is also affected in gukh mutants. In fact, considering the hypomorphic character of the gukh alleles that were used in this study, the effect on Scrib localization appears remarkably strong. This observation might indicate that GUKh activity is required only temporarily and/or in a locally restricted fashion to prime a secondary mechanism by which Scrib becomes associated with the SSR, e.g., through a more direct interaction with Dlg. Interestingly, presynaptic expression of GUKh-C is largely sufficient to restore postsynaptic Scrib localization at gukh mutant NMJs. Together, these observations suggest a second, more indirect mechanism by which GUKh contributes to the recruitment of Scrib to the postsynaptic SSR; such a mechanism may involve trans-synaptic signaling (Mathew, 2002).

These studies provide evidence for one mechanism by which scaffolding proteins with different interaction domains may be linked to form a network of multiprotein complexes. GUKh, in physically linking Dlg and Scrib, can therefore bring together these complexes and their associated proteins. Since a single protein forms this link, it would be a straightforward point at which to also separate the complexes, along with their actions, to regulate different aspects of synapse formation. Examples would be during synapse stabilization and during synapse growth through bouton budding. Thus, this work provides a means by which macromolecular complexes can mediate and finely tune various structural changes at the highly dynamic structure of the synapse (Mathew, 2002).

Polarized cells contain numerous membrane domains, but it is unclear how the formation of these domains is coordinated to create a single integrated cell architecture. Genetic screens of Drosophila embryos have identified three complexes, each containing one of the PDZ domain proteins -- Stardust (Sdt), Bazooka (Baz) and Scribble (Scrib) -- that control epithelial polarity and formation of zonula adherens. These complexes can be ordered into a single regulatory hierarchy that is initiated by cell adhesion-dependent recruitment of the Baz complex to the zonula adherens. The Scrib complex represses apical identity along basolateral surfaces by antagonizing Baz-initiated apical polarity. The Sdt-containing Crb complex is recruited apically by the Baz complex to counter antagonistic Scrib activity. Thus, a finely tuned balance between Scrib and Crb complex activity sets the limits of the apical and basolateral membrane domains and positions cell junctions. These data suggest a model in which the maturation of epithelial cell polarity is driven by integration of the sequential activities of PDZ-based protein complexes (Bilder, 2003).

Domain mapping of Scrib reveals a multistep localization mechanism and domains necessary for establishing cortical polarity

The Drosophila tumor suppressor protein Scribble is required for epithelial polarity, neuroblast polarity, neuroblast spindle asymmetry and limiting cell proliferation. It is a member of the newly described LAP protein family, containing 16 leucine rich repeats (LRRs), four PDZ domains and an extensive carboxyl-terminal (CT) domain. LRR and PDZ domains mediate protein-protein interactions, but little is know about their function within LAP family proteins. This study has determined the role of the LRR, PDZ and CT domains for Scribble localization in neuroblasts and epithelia, and for Scribble function in neuroblasts. It was found that the LRR and PDZ domains are both required for proper targeting of Scribble to septate junctions in epithelia; that the LRR domain is necessary and sufficient for cortical localization in mitotic neuroblasts, and that the PDZ2 domain is required for efficient cortical and apical localization of Scribble in neuroblasts. In addition, it is shown that the LRR domain is sufficient to target Miranda protein to the neuroblast cortex, but that LRR+PDZ will exclude Miranda from the cortex. These results highlight the importance of both LRR and PDZ domains for the proper localization and function of Scribble in neuroblasts (Albertson, 2004).

The results demonstrate that the Scrib LRRs are absolutely required for Scrib cortical localization in neuroblasts and epithelial cells. How might the LRRs direct Scrib to the plasma membrane? Little is known about binding specificities of LRRs in LAP proteins, yet LRRs in other cytoplasmic, transmembrane and extracellular proteins have been extensively studied. One LRR subfamily has an LRR-containing extracellular domain that is responsible for high-affinity binding to ligands. Another LRR protein subfamily binds small cytoplasmic GTPases, such as Ras and Ran. Based on these reports, activated small G proteins (Cdc42, Rac1, Ran) are candidates for cortical targeting of the Scrib LRR domain, although the distribution of these proteins in neuroblasts has not been determined, nor have they been tested for physical interactions with Scrib LRRs (Albertson, 2004).

Scrib proteins lacking all four PDZ domains, or just PDZ2, are detected in the cytoplasm and uniformly around the neuroblast cortex, with only occasional weak apical enrichment. Scrib PDZ2 is likely to target Scrib to the neuroblast cortex, and more specifically to the apical neuroblast cortex, by associating with Gukh and Dlg proteins. Gukh binds to both Scrib PDZ2 and Dl; Scrib, Gukh and Dlg are all co-localized at the neuroblast cortex; dlg mutants have cytoplasmic Scrib protein. One simple model consistent with these data is that Dlg binds Gukh, which binds Scrib PDZ2, resulting in the observed localization of Scrib within neuroblasts. Nevertheless, the PDZ domains are not sufficient for proper Scrib localization: Scrib proteins that contain the PDZs but lack the LRRs (DeltaLRR or PDZ) are completely cytoplasmic. A two-step model is proposed for Scrib localization in which Scrib LRR-dependent cortical localization precedes and is a prerequisite for Scrib PDZ2-dependent apical enrichment. It is noted that proteins lacking all four PDZ domains still show weak apical enrichment in some neuroblasts, however, indicating a minor role for the LRR domain in apical Scrib targeting (Albertson, 2004).

The data suggest a mechanism in which Scrib neuroblast and epithelial localization involves distinct steps, moving from cytoplasm to cortex then apically. Other proteins targeted to specific cortical domains within neuroblasts show a similar cortical-to-asymmetric localization mechanism. The Insc protein contains a 158 aa region that directs Insc to the neuroblast cortex; the addition of a separate 100 aa domain confers apical localization. Similarly, the Pins protein contains four C-terminal GoLoco repeats that are sufficient for cortical localization, but addition of the first three of seven N-terminal tetratricopeptide repeats (TPRs) is required for asymmetric apical targeting. Stepwise targeting of Dlg has also been observed in epithelia and neuromuscular junctions. In epithelia, the Hook domain targets Dlg to the cell cortex while PDZ2 is required to restrict Dlg to the septate junction. At neuromuscular junctions, the Hook domain is necessary to localize Dlg to the plasma membrane and both PDZ1 and PDZ2 domains promote transport to the synapse. These observations suggest cortical localization may be a prerequisite for subsequent targeting to specific membrane domains and raise the possibility that similar transport mechanisms are shared among protein complexes in neuroblasts, epithelia and synapses (Albertson, 2004).

Despite the fact that Scrib has a plethora of protein-protein interaction motifs, only one binding partner for Drosophila Scrib has been identified, Gukh, which binds to Scrib PDZ2. Binding assays such as yeast two-hybrid assays and mass spectrometry will be essential to identify additional Drosophila Scrib binding partners, as has been shown for mammalian LAP proteins. Such discoveries will enable the determination of upstream and downstream players in Scrib-mediated pathways and will further an understanding of many cellular functions, including establishment of epithelial cell polarity, stem cell division, mitotic spindle biology, cell cycle progression and synapse formation and homeostasis (Albertson, 2004).

Scrib proteins lacking LRRs fail to be properly targeted to the lateral membrane and septate junctions of mature epithelial cells, whereas Scrib proteins lacking all four PDZ domains, or just PDZ2, still show some septate junction enrichment with an elevated level of cytoplasmic protein. Thus, the LRR domains are the primary determinant of cortical/septate junction targeting, and PDZ2 increases the efficiency or stability of this localization. Scrib localization in epithelia also requires Dlg, but the role of the Dlg and Scrib PDZ2-binding protein Gukh have not been examined in epithelia. The ability of each Scrib domain to establish or maintain epithelial polarity could not be tested in this system, however, because zygotic scrib mutant embryos develop normal epithelial polarity because of maternally derived Scrib protein (even though there is no detectable maternal Scrib protein in stage 15 or later embryos) (Albertson, 2004).

The proteins that interact with the Scrib LRRs to mediate cortical association in epithelia are unknown. More is known about the role of PDZ domains in junctional targeting of LAP proteins. The mammalian LAP proteins Erbin and Densin180 show PDZ-mediated interactions with p120-catenins, and Erbin-catenin p0071 colocalize to adherens junctions and desmosomes in cultured epithelial cells. Disruption of Erbin-p0071 interactions leads to aberrant cell morphology and disruption of cell-cell contacts. Similarly, Drosophila Scrib is required for proper morphology and formation of septate junctions in epithelia, and the PDZ domains are necessary for efficient Scrib localization to the epithelial cell septate junctions. Thus, Drosophila catenins are excellent candidates for recruiting Scrib to the septate junction (Albertson, 2004 and references therein).

In the absence of all Scrib function, Mira is predominantly localized to the cytoplasm and mitotic spindle of neuroblasts. Expression of just the Scrib LRR domain results in uniform cortical Scrib LRR distribution and the restoration of uniform cortical Mira localization. Conversely, all Scrib proteins that lack the LRR domain (PDZ, DeltaLRR and CT) fail to efficiently target Mira to the cortex. These results reveal a positive role for the Scrib LRR domain in targeting Mira to all regions of the neuroblast cortex. A similar 'uniform cortical Mira' phenotype is also observed in certain aPKC and lgl genetic backgrounds. Neuroblasts lacking aPKC show uniform cortical Mira and neuroblasts misexpressing a dephospho-Lgl protein also show uniform cortical Mira. In addition, loss of lgl leads to cytoplasmic Mira localization in neuroblasts. This has led to a model in which the apically-localized aPKC phosphorylates Lgl to inactivate it, thus restricting active dephospho-Lgl to the basal cortex, where it promotes cortical localization of Mira. The Scrib LRRs could act upstream of aPKC and Lgl, perhaps by blocking aPKC/Lgl interactions, and thus allowing activated Lgl to target Mira to the entire cortex. Alternatively, the Scrib LRRs could act downstream of aPKC and Lgl, perhaps by allowing both dephospho- and phospho-Lgl to target Mira to the cortex. In addition, loss of jaguar (myosin VI) leads to cytoplasmic localization of Mira, raising the possibility that the Scrib LRRs could stimulate myosin VI activity around the neuroblast cortex to promote uniform cortical Mira localization. The identification of Scrib LRR-binding proteins will help distinguish between these models (Albertson, 2004).

Addition of the PDZ domains back to the Scrib LRR protein dramatically alters the function of the protein. Whereas the Scrib LRR protein is uniformly cortical and promotes Mira cortical localization, Scrib LRR+PDZ proteins (FL, DeltaCT) are apically enriched and exclude Mira from the apical cortex. Thus, addition of the PDZ domains switches Scrib from promoting cortical Mira localization to excluding cortical Mira localization. The PDZ domains could carry out this function of excluding Mira from the apical cortex in at least three different ways. (1) The Scrib PDZ domains could promote aPKC-Lgl interactions, thereby leading to the phosphorylation and inactivation of apical Lgl; this would restrict active Lgl to the basal cortex, where it promotes cortical Mira localization. (2) The Scrib PDZ domains could promote myosin II (zipper) activity at the apical cortex; myosin II is a known inhibitor of Lgl, and thus this would restrict active Lgl to the basal cortex where it could promote cortical Mira localization. (3) The Scrib PDZ domains could provide directionality to the actin-myosin VI cytoskeleton, which could transport Mira specifically to the basal cortex. Identification of proteins that interact with the Scrib PDZ domains would help distinguish between these models (Albertson, 2004).

Little is known about how Dlg, Scrib and Lgl regulate cell size asymmetry and spindle asymmetry. LRR and PDZ domains of Scrib are necessary for this function. How might Scrib regulate cell size and spindle asymmetry? Two good candidate effectors are Ran GTPase and Pins. LRRs are known to physically interact with Ran, which promotes spindle assembly through several target proteins. For example, Ran stimulates the activity of NuMA (a microtubule motor accessory protein that promotes spindle assembly) by destabilizing inhibitory complexes associated with NuMA. LGN (a mammalian Pins homolog) is essential for mitotic spindle assembly and binds NuMA; release from LGN is an important event in the activation of mitotic NuMA. In Drosophila, Pins physically interacts with Dlg and is asymmetrically localized to the apical cortex of mitotic neuroblasts, where it promotes spindle asymmetry. These data suggest possible links between Dlg, Scrib, Ran and Pins and establishment of mitotic spindle asymmetry. Genetic and biochemical studies investigating interactions between Scrib, Ran and Pins may further understanding of spindle asymmetry establishment in Drosophila neuroblasts (Albertson, 2004).

Deletion of the CT domain has no effect on Scrib localization or its ability to rescue all tested scrib mutant phenotypes in neuroblasts; the CT domain alone is cytoplasmic and has no rescuing ability in any assay performed. It is concluded that the CT domain is not essential for any aspect of Scrib localization or function tested here (Albertson, 2004).

scrib transgenic lines that specifically lack the LAPSDa/b domains have not been assayed, however, some conclusions can be drawn based on existing Scrib domain analysis. The Scrib DeltaPDZ protein contains both LAPSDa/b domains, is membrane targeted but not enriched apically; it fails to promote basal Mira targeting, and it is defective for asymmetric mitotic spindle and cell size asymmetry. Thus, the LAPSD domains are insufficient for apical enrichment of Scrib and all of its tested functions in neuroblasts. The Scrib LRR protein lacking the LAPSDb domain is still membrane-associated, showing that the LAPSDb domain is not required for Scrib membrane targeting. Some evidence was found that the LAPSDb domain regulates nuclear import/export of the Scrib protein. The LRR protein contains just the LRRs and the LAPSDa domain and is targeted to the nucleus; this shows that there is a nuclear import signal or binding site for a nuclear protein within the LRR/LAPSDa domains, although a predicted nuclear localization signal is not detectable within these domains. In contrast, the DeltaPDZ protein contains the same LRR/LAPSDa domains plus the LAPSDb and CT domains, and it is excluded from the nucleus. This shows that the LAPSDb or CT domains can prevent nuclear import of the LRR/LAPSDa protein; it is highly likely that this function is provided by the LAPSDb domain, because deletion of the CT domain from an otherwise wild-type Scrib protein (i.e., DeltaCT) does not result in nuclear localization. These results are in contrast to the role of the LAPSDa/b domains in the related C. elegans Let-413 protein, where the LAPSDa/b domains are required for establishing epithelial polarity but not Let-413 protein localization. It will be interesting to determine whether the Scrib LAPSDa/b domains play a similar role in Scrib epithelial localization (Albertson, 2004).

The leading edge during dorsal closure as a model for epithelial plasticity: Pak is required for recruitment of the Scribble complex and septate junction formation

Dorsal closure (DC) of the Drosophila embryo is a model for the study of wound healing and developmental epithelial fusions, and involves the sealing of a hole in the epidermis through the migration of the epidermal flanks over the tissue occupying the hole, the amnioserosa. During DC, the cells at the edge of the migrating epidermis extend Rac- and Cdc42-dependent actin-based lamellipodia and filopodia from their leading edge (LE), which exhibits a breakdown in apicobasal polarity as adhesions are severed with the neighbouring amnioserosa cells. Studies using mammalian cells have demonstrated that Scribble (Scrib), an important determinant of apicobasal polarity that functions in a protein complex, controls polarized cell migration through recruitment of Rac, Cdc42 and the serine/threonine kinase Pak, an effector for Rac and Cdc42, to the LE. DC and the follicular epithelium were used to study the relationship between Pak and the Scrib complex at epithelial membranes undergoing changes in apicobasal polarity and adhesion during development. It is proposed that, during DC, the LE membrane undergoes an epithelial-to-mesenchymal-like transition to initiate epithelial sheet migration, followed by a mesenchymal-to-epithelial-like transition as the epithelial sheets meet up and restore cell-cell adhesion. This latter event requires integrin-localized Pak, which recruits the Scrib complex in septate junction formation. It is concluded that there are bidirectional interactions between Pak and the Scrib complex modulating epithelial plasticity. Scrib can recruit Pak to the LE for polarized cell migration but, as migratory cells meet up, Pak can recruit the Scrib complex to restore apicobasal polarity and cell-cell adhesion (Bahri, 2010).

Some embryos lacking zygotic Pak function successfully bring the epidermal flanks together at the dorsal midline but fail to restore septate junctions and adherens junctions at the LE in the DME cells. Thus, Pak at the LE membrane of the dorsal-most epithelial cells (DME) is regulating establishment of apicobasal polarity during a mesenchymal-epithelial transition. It is suspected that Pak is acting through different routes in its regulation of adherens junction formation versus septate junction formation. This study has focused on Pak regulation of the Scrib complex in septate junction formation at the LE. The data indicate that Pak is a component of the Scrib complex at the lateral membrane. Although Pak might be associating with the Scrib complex throughout epithelia, it might only be required for recruitment of the Scrib complex in epithelia derived from a mesenchymal-like intermediate such as the follicular epithelium and the LE. With the exception of the LE, apicobasal polarity in the epidermis is determined much earlier in development with formation of the blastoderm by cellularization. The epidermis is therefore a primary epithelium that does not arise from a mesenchymal intermediate, and Pak function does not appear to be required for apicobasal polarity in primary epithelia (Bahri, 2010).

Localization of Pak at the lateral membrane in both the follicular epithelium and in the epidermis is integrin-dependent. Studies using organ culture of embryonic kidney mesenchyme and MDCK cells demonstrate a requirement for integrins in apicobasal polarity of epithelia derived from MET, and this study has shown that βPS-integrin is required for Scrib complex and septate junction protein recruitment at the LE and in the follicular epithelium. Furthermore, previous studies in the follicular epithelium and another Drosophila epithelium derived from MET, the midgut, have demonstrated a requirement for integrins in the maintenance of apicobasal polarity. It is proposed that, at the LE, the absence of the septate junction diffusion barrier allows the accumulation of integrin complexes along the lateral membrane. These lateral integrin complexes recruit Pak, around which the Scrib complex is assembled. Thus, the absence of septate junctions allows the recruitment of proteins needed for the assembly of septate junctions. The model suggests that there might be transient Pak-mediated links between integrin and the Scrib complex. Interestingly, Dlg and βPS-integrin have been shown to co-immunoprecipitate from fly head extracts, consistent with these proteins existing in a complex in the nervous system and/or in epithelia (Bahri, 2010).

The data and recent studies on the amnioserosa support the idea that septate junctions restrict the accumulation of lateral integrins. The amnioserosa is devoid of septate junction proteins such as FasIII, and this might be owing to absence in this tissue of the transcription factor Grainy head, which promotes expression of septate junction proteins. The wild-type amnioserosa has high levels of lateral βPS-integrin, but ectopic expression of Grainy head in the amnioserosa leads to an accumulation of septate junction proteins and an accompanying disruption of βPS-integrin localization. Similarly, at the completion of DC, septate junctions appear at the LE and this is accompanied by downregulation of LE lateral integrins. In pak14pak376A and cora14 embryos where LE septate junctions are deficient, lateral LE βPS-integrin persists (Bahri, 2010).

A recent study in mammalian cell culture indicates that Scrib recruits Pak to the LE (Nola, 2008), and this study has shown that Pak localization in the follicular epithelium is Scrib-dependent. This study of the LE at the end of DC demonstrates that the relationship between Cdc42/Rac signaling complexes and Scrib can act in the opposite direction: membrane-localized Pak recruits the Scrib complex. A bidirectional interaction between the Scrib complex and Cdc42/Rac signaling complexes, including Pak, might be a crucial regulator of events at the LE of closing epithelia during both wound healing and development in diverse systems. Scrib at the newly formed LE can lead to recruitment of the Cdc42/Rac signaling complex, allowing acquisition of mesenchymal characteristics and polarized cell migration. When the opposing epithelial flanks meet up, events can be reversed with Pak recruiting the Scrib complex to the lateral membrane, contributing to restoration of apicobasal polarity and cell adhesion at the LE during MET. The Scrib/Pak complex is believed to be a 'toggle switch', enabling the epithelial membrane to shift back and forth between a migratory state characterized by actin-based extensions and an apicobasal polarized state characterized by cell-cell adhesion (Bahri, 2010).

Aurora A triggers Lgl cortical release during symmetric division to control planar spindle orientation

Mitotic spindle orientation is essential to control cell-fate specification and epithelial architecture. The tumor suppressor Lgl localizes to the basolateral cortex of epithelial cells, where it acts together with Dlg and Scrib to organize apicobasal polarity. Dlg and Scrib also control planar spindle orientation but how the organization of polarity complexes is adjusted to control symmetric division is largely unknown. Lgl redistribution during epithelial mitosis is reminiscent of asymmetric cell division, where it is proposed that Aurora A promotes aPKC activation to control the localization of Lgl and cell-fate determinants. This study shows that the Dlg complex is remodeled during Drosophila follicular epithelium cell division, when Lgl is released to the cytoplasm. Aurora A controlled Lgl localization directly, triggering its cortical release at early prophase in both epithelial and S2 cells. This relied on double phosphorylation within the putative aPKC phosphorylation site, which was required and sufficient for Lgl cortical release during mitosis and could be achieved by a combination of aPKC and Aurora A activities. Cortical retention of Lgl disrupted planar spindle orientation, but only when Lgl mutants that could bind Dlg were expressed. Taken together, Lgl mitotic cortical release is not specifically linked to the asymmetric segregation of fate determinants, and the study proposes that Aurora A activation breaks the Dlg/Lgl interaction to allow planar spindle orientation during symmetric division via the Pins (LGN)/Dlg pathway (Carvalho, 2015).

Evolutionarily conserved polarity complexes establish distinct membrane domains and the polarized assembly of junctions along the apicobasal axis has been extensively characterized. One general feature is that it relies on mutual antagonism between apical atypical protein kinase C (aPKC) and Crumbs complexes and a basolateral complex formed by Scribble (Scrib), Lethal giant larvae (Lgl), and Discs large (Dlg). This study used the Drosophila follicular epithelium as an epithelial polarity model to address how polarity is coordinated during symmetric division. Dlg and Scrib have been shown to provide a lateral cue for planar spindle orientation. Accordingly, Scrib and Dlg remain at the cortex during follicle cell division. In contrast, Lgl is released from the lateral cortex to the cytoplasm during mitosis. This subcellular reallocation begins during early prophase, since Lgl starts to be excluded from the cortex prior to cell rounding, one of the earliest mitotic events, and is completely cytoplasmic before nuclear envelope breakdown (NEB). Thus, the Dlg complex is remodeled at mitosis onset in epithelia (Carvalho, 2015).

The subcellular localization of Lgl is controlled by aPKC-mediated phosphorylation of a conserved motif, which blocks Lgl interaction with the apical cortex. To address the mechanism of cortical release during mitosis, nonphosphorytable form Lgl3A-GFP was expressed in the follicular epithelium. Lgl3A-GFP remains at the cortex throughout mitosis indicating that Lgl dynamics during epithelial mitosis also rely on the aPKC phosphorylation motif. Although the apical aPKC complex depolarizes during follicle cell division, Lgl cortical release precedes aPKC depolarization. Using Par-6-GFP as a marker for the aPKC complex and the Lgl cytoplasmic accumulation as readout of its cortical release, it was found that maximum cytoplasmic accumulation of Lgl occurs when most Par-6 is still apically localized (~70% relative to interphase levels). Thus, Lgl cortical release is the first event of the depolarization that characterizes follicle cell division, indicating that Lgl reallocation does not require extension of aPKC along the lateral cortex (Carvalho, 2015).

Although the major pools of Lgl and aPKC are segregated during interphase, Lgl has a dynamic cytoplasmic pool that rapidly exchanges with the cortex. Thus, further activation of aPKC at mitosis onset would be expected to shift the equilibrium toward cytoplasmic localization. Lgl dynamic redistribution in epithelia is similar to the neuroblast, where activation of Aurora A (AurA) leads to Par-6 phosphorylation and subsequent aPKC activation. To test whether a similar mechanism induced Lgl cortical release during epithelial mitosis, Lgl subcellular localization was analyzed in aPKC mutants and in par-6 mutants unphosphorylatable by AurA. Lgl cytoplasmic accumulation is unaffected in par-6; par-6S34A mutant cells. Temperature-sensitive aPKCts/aPKCk06403 mutants display strong cytoplasmic accumulation of Lgl during prophase, with a minor delay relatively to the wild-type). Moreover, homozygous mutant clones for null (aPKCk06403) and kinase-defective (aPKCpsu141) alleles also display Lgl cortical release during mitosis. These results implicate that although aPKC activity may contribute for Lgl mitotic dynamics, the putative aPKC phosphorylation motif is under the control of a different kinase, which triggers Lgl cortical release in the absence of aPKC (Carvalho, 2015).

AurA is a good candidate to induce Lgl cortical release as it controls polarity during asymmetric division. Furthermore, Drosophila AurA is activated at the beginning of prophase, which coincides with the timing of Lgl cytoplasmic reallocation. To examine whether AurA controls Lgl dynamics in the follicular epithelium, homozygous mutant clones were generated for the kinase-defective allele aurA37. In contrast to wild-type cells, only low amounts of cytoplasmic Lgl were detected during prophase in aurA37 mutants, which display a pronounced delay in the cytoplasmic reallocation of Lgl during mitosis. This delayed cortical release of Lgl has been previously reported during asymmetric cell division in aurA37 mutants, possibly resulting from residual kinase activity. Thus, AurA is essential to trigger Lgl cortical exclusion at epithelial mitosis onset (Carvalho, 2015).

The idea that Lgl mitotic reallocation is directly controlled by a mitotic kinase implies that Lgl should display similar dynamics regardless of the polarized status of the cell. Consistently, Lgl-GFP is also released from the cortex before NEB in nonpolarized Drosophila S2 cells. Furthermore, Lgl3A-GFP is retained in the cortex during mitosis, revealing that Lgl cortical release is also phosphorylation dependent in S2 cells. Treatment with a specific AurA inhibitor (MLN8237), or with aurA RNAi, strongly impairs Lgl cortical release during prophase, as Lgl is present in the cortex at NEB. However, inhibition of AurA still allows later cortical exclusion, which could result from the activity of another kinase. Despite their distinct roles, AurA and Aurora B (AurB) phosphorylate common substrates in vitro. Therefore, whether AurB could act redundantly with AurA was analyzed. Inactivation of AurB with a specific inhibitor, Binucleine 2, enables normal Lgl cytoplasmic accumulation before NEB and still allows later cortical exclusion in cells treated simultaneously with the AurA inhibitor As AurB does not seem to participate on Lgl mitotic dynamics, RNAi directed against aPKC was used to examine whether it could act redundantly with AurA. aPKC depletion did not block Lgl cortical exclusion, but it was slightly delayed. However, simultaneous AurA inhibition and aPKC RNAi produced almost complete cortical retention of Lgl during mitosis. Thus, AurA induces Lgl release during early prophase, but aPKC retains its ability to phosphorylate Lgl during mitosis (Carvalho, 2015).

To address which serine(s) within the phosphorylation motif of Lgl control its dynamics during mitosis, individual and double mutants were enerated. As complete cortical release occurs before NEB, the ratio of cytoplasmic to cortical mean intensity of Lgl-GFP at NEB was quantified to compare each different mutant. All the single mutants displayed similar dynamics to LglWT, exiting to the cytoplasm prior to NEB. In contrast, all double mutants were cortically retained during mitosis, indicating that double phosphorylation is both sufficient and required to efficiently block Lgl cortical localization (Carvalho, 2015).

The ability to doubly phosphorylate Lgl would explain how AurA drives Lgl cortical release. Accordingly, the sequence surrounding S656 perfectly matches AurA phosphorylation consensus, whereas the S664 surrounding sequence shows an exception in the -3 position. In contrast, the sequence surrounding S660 does not resemble AurA phosphorylation consensus, and AurA does not directly phosphorylate S660 in vitro as detected by phosphospecific antibodies against S660. That S656 is directly phosphorylated by recombinant AurA was confirmed in vitro using a phosphospecific antibody for S656. Moreover, AurA inhibition or aurA RNAi results in a similar cortical retention at NEB to LglS656A,S664A, suggesting that AurA also controls S664 phosphorylation during mitosis, whereas aPKC would be the only kinase active on S660. Consistent with this, aPKC RNAi increases the cortical retention of LglS656A,S664A, mimicking the localization of Lgl3A. Furthermore, whereas S660A mutation does not significantly affect the cytoplasmic accumulation of Lgl in aPKC RNAi, S656A and S664A mutations disrupt Lgl cortical release in aPKC-depleted cells, leading to the degree of cortical retention of LglS656A,S660A and LglS660A,S664A, respectively. Altogether, these results support that AurA controls S656 and S664 and that these phosphorylations are partially redundant with aPKC phosphorylation to produce doubly phosphorylated Lgl, which is released from the cortex (Carvalho, 2015).

RNAi-mediated knockdown of Lgl in vertebrate HEK293 cells results in defective chromosome segregation. Furthermore, overexpressed Lgl-GFP shows a slight enrichment on the mitotic spindle suggesting that relocalization of Lgl could be important to control chromosome segregation. However, lgl mutant follicle cells assemble normal bipolar spindles, and although it was possible to detect minor defects on chromosome segregation, the mitotic timing (time between NEB and anaphase) is indistinguishable between lgl and wild-type cells. Additionally, loss of Lgl activity allows proper chromosome segregation in both Drosophila S2 cells and syncytial embryos. Thus, Lgl does not seem to have a general role in the control of faithful chromosome segregation in Drosophila (Carvalho, 2015).

Nevertheless, Lgl cortical release could per se play a mitotic function, as key mitotic events are controlled at the cortex. In fact, the orientation of cell division requires the precise connection between cortical attachment sites and astral microtubules, which relies on the plasma membrane associated protein Pins (vertebrate LGN). Pins uses its TPR repeat domain to bind Mud (vertebrate NUMA), which recruits the dynein complex to pull on astral microtubules, and its linker domain to interact with Dlg, which participates on the capture of microtubule plus ends. Notably, Pins/LGN localizes apically during interphase in Drosophila and vertebrate epithelia, being reallocated to the lateral cortex to orient cell division. Pins relocalization relies on aPKC in some epithelial tissues, but not in chick neuroepithelium and in the Drosophila follicular epithelium, where Dlg provides a polarity cue to restrict Pins to the lateral cortex. Dlg controls Pins localization during both asymmetric and symmetric division, and a recent study has shown that vertebrate Dlg1 recruits LGN to cortex via a direct interaction. However, Dlg uses the same phosphoserine binding region within its guanylate kinase (GUK) domain to interact with Pins/LGN and Lgl. Thus, maintenance of a cortical Dlg/Lgl complex during mitosis is expected to impair the ability of Dlg to bind Pins and control spindle orientation (Carvalho, 2015).

Interaction between the Dlg's GUK domain and Lgl requires phosphorylation of at least one serine within the aPKC phosphorylation site. Although the phosphorylation-dependent binding of Lgl to Dlg remains to be shown in Drosophila, crystallographic studies revealed that all residues directly involved in the interaction with p-Lgl are evolutionarily conserved from C. elegans to humans. Thus, whereas Lgl3A does not form a fully functional Dlg/Lgl polarity complex, double mutants should bind Dlg's GUK domain and are significantly retained at the cortex during mitosis due to the inability to be double phosphorylated. This led to an examination of their ability to support epithelial polarization during interphase and to interfere with mitotic spindle orientation. Rescue experiments were performed in mosaic egg chambers containing lgl27S3 null follicle cell clones. lgl mutant clones display multilayered cells with delocalization of aPKC. This phenotype is rescued by Lgl-GFP, but not by Lgl3A-GFP. More importantly, in contrast to LglS660A,S664A, which extends to the apical domain in wild-type cells and fails to rescue epithelial polarity in lgl mutant cells, LglS656A,S660A and LglS656A,S664A can rescue epithelial polarity, localizing with Dlg at the lateral cortex and below aPKC. Hence, aPKC-mediated phosphorylation of S660 or S664 is sufficient on its own to control epithelial polarity and to confine Lgl to the lateral cortex (Carvalho, 2015).

Whether exclusion of Lgl from the cortex and the consequent release from Dlg would be functionally relevant for oriented cell division was examined. Expression of Lgl-GFP or Lgl3A-GFP does not affect planar spindle orientation during follicle cell division. In contrast, Lgl double mutants display metaphasic cells in which the spindle axis, determined by centrosome position, is nearly perpendicular to the epithelial layer. Live imaging revealed that these spindle orientation defects were maintained throughout division as it was possible to follow daughter cells separating along oblique and perpendicular angles to the epithelia. Moreover, equivalent defects on planar spindle orientation were detected upon expression of LglS656A,S664A in the lgl or wild-type background, indicating that cortical retention of Lgl exerts a dominant effect. Interestingly, LglS656A,S660A and LglS656A,S664A induce higher randomization of angles, whereas LglS660A,S664A, which is less efficiently restricted to the lateral cortex, produces a milder phenotype. Altogether, these results indicate that retention of Lgl at the lateral cortex disrupts planar spindle orientation only if Lgl can interact with Dlg (Carvalho, 2015).

Despite the ability of LglS656A,S660A-GFP to rescue epithelial polarity in lgl mutants, strong overexpression of LglS656A,S660A-GFP, but not of other Lgl double mutants, can dominantly disrupt epithelial polarity during the proliferative stages of oogenesis. One interpretation is that LglS656A,S660A forms the most active lateral complex of the mutant transgenes, disrupting the balance between apical and lateral domains. Therefore whether the dominant effect of Lgl cortical retention on spindle orientation could solely result from Dlg mislocalization was assessed. Dlg is properly localized at the lateral cortex in LglS656A,S660A-expressing cells presenting misoriented spindles, but this position does not correlate with the orientation of the centrosomes. Thus, cortical retention of Lgl interferes with Dlg's ability to transmit its lateral cue to instruct spindle orientation, which may result from an impairment of the Dlg/Pins interaction (Carvalho, 2015).

In conclusion, these findings outline a mechanism that explains how the lateral domain is remodeled to accomplish oriented epithelial cell division, unveiling that AurA has a central role in controlling the subcellular distribution of Lgl. AurA regulates the activity of aPKC at mitotic entry during asymmetric division, and these results are consistent with the ability of aPKC to phosphorylate and collaborate in Lgl cortical release. However, in epithelia, aPKC accumulates in the apical side during interphase, where it induces apical exclusion of Lgl, in part by generating a phosphorylated form that binds Dlg. Consequently, aPKC has a reduced access to the cortical pool of Lgl at mitotic entry and would be unable to rapidly induce Lgl cortical exclusion. These data show that cell-cycle-dependent activation of AurA removes Lgl from the lateral cortex through AurA's ability to control Lgl phosphorylation on S656 and S664 independently of aPKC. Thus, AurA and aPKC exert the spatiotemporal control of Lgl distribution to achieve unique cell polarity roles in distinct cell types (Carvalho, 2015).

It is proposed that release of Lgl from the cortex allows Dlg interaction with Pins to promote planar cell division in Drosophila epithelia. Lgl cortical release requires double phosphorylation, indicating that whereas Lgl-Dlg association involves aPKC phosphorylation, multiple phosphorylations break this interaction, acting as an off switch on Lgl-Dlg binding. Triple phosphomimetic Lgl mutants display weak interactions with Dlg, suggesting that multiple phosphorylations could directly block Lgl-Dlg interaction. Alternatively, the negative charge of two phosphate groups may suffice to induce association between the N- and C-terminal domains of Lgl, impairing its ability to interact with the cytoskeleton and plasma membrane as previously proposed. This would reduce the local concentration of Lgl available to interact with Dlg, enabling the interaction of Dlg's GUK domain with the pool of Pins phosphorylated by AurA. Therefore, AurA converts the Lgl/Dlg polarity complex generated upon aPKC phosphorylation into the Pins/Dlg spindle orientation complex. This study, underlines the critical requirement of synchronizing the cell cycle with the reorganization of polarity complexes to achieve precise control of spindle orientation in epithelia (Carvalho, 2015).

Scribbled optimizes BMP signaling through its receptor internalization to the Rab5 endosome and promote robust epithelial morphogenesis

Epithelial cells are characterized by apical-basal polarity. Intrinsic factors underlying apical-basal polarity are crucial for tissue homeostasis and have often been identified to be tumor suppressors. Patterning and differentiation of epithelia are key processes of epithelial morphogenesis and are frequently regulated by highly conserved extrinsic factors. However, due to the complexity of morphogenesis, the mechanisms of precise interpretation of signal transduction as well as spatiotemporal control of extrinsic cues during dynamic morphogenesis remain poorly understood. Wing posterior crossvein (PCV) formation in Drosophila serves as a unique model to address how epithelial morphogenesis is regulated by secreted growth factors. Decapentaplegic (Dpp), a conserved bone morphogenetic protein (BMP)-type ligand, is directionally trafficked from longitudinal veins (LVs) into the PCV region for patterning and differentiation. These data reveal that the basolateral determinant Scribbled (Scrib) is required for PCV formation through optimizing BMP signaling. Scrib regulates BMP-type I receptor Thickveins (Tkv) localization at the basolateral region of PCV cells and subsequently facilitates Tkv internalization to Rab5 endosomes, where Tkv is active. BMP signaling also up-regulates scrib transcription in the pupal wing to form a positive feedback loop. These data reveal a unique mechanism in which intrinsic polarity genes and extrinsic cues are coupled to promote robust morphogenesis (Gui, 2016).

This study shows that the Scrib complex, a basolateral determinant, is a novel feedback component that optimizes BMP signaling in the PCV region of the Drosophila pupal wing (Gui, 2016).

During PCV development, limited amounts of Dpp ligands are provided by the Dpp trafficking mechanism. Furthermore, amounts of receptors appear to be limited since tkv transcription is down-regulated in the cells in which the BMP signal is positive, a mechanism that serves to facilitate ligand diffusion and sustain long-range signaling in the larval wing imaginal disc. To provide robust signal under conditions in which both ligands and receptors are limiting, additional molecular mechanisms are needed. Previous studies suggest that two molecules play such roles. Crossveinless-2 (Cv-2), which is highly expressed in the PCV region, serves to promote BMP signaling through facilitating receptor-ligand binding. Additionally, the RhoGAP protein Crossveinless-c (Cv-c) provides an optimal extracellular environment to maintain ligand trafficking from LVs into PCV through down-regulating integrin distribution at the basal side of epithelia. Importantly, both cv-2 and cv-c are transcriptionally regulated by BMP signaling to form a feedback or feed-forward loop for PCV formation (Gui, 2016).

Scrib, a third component, sustains BMP signaling in the PCV region in a different manner. First, to utilize Tkv efficiently, Scrib regulates Tkv localization at the basolateral region in the PCV cells, where ligand trafficking takes place. Regulation of receptor localization could be a means of spatiotemporal regulation of signaling molecules during the dynamic process of morphogenesis. Second, to optimize the signal transduction after receptor-ligand binding, Scrib facilitates Tkv localization in the Rab5 endosomes. Localization of internalized Tkv is abundant at Rab5 endosomes in the PCV region of wild-type, but not scrib RNAi cells. While the physical interaction between Scrib, Tkv and Rab5 in the pupal wing remains to be addressed, the data in S2 cells suggest that physical interactions between these proteins are key for preferential localization of Tkv at the Rab5 endosomes. Recently, Scrib has been implicated in regulating NMDA receptor localization through an internalization-recycling pathway to sustain neural activity. Therefore, Scrib may be involved in receptor trafficking in a context-specific manner, the molecular mechanisms of which, however, remain to be characterized. Third, BMP/Dpp signaling up-regulates scrib transcription in the pupal wing. Moreover, knockdown of scrib leads to loss of BMP signaling in PCV region but not loss of apical-basal polarity. These facts suggest that upregulation of Scrib is key for optimizing BMP signaling by forming a positive feedback loop (Gui, 2016).

Previous studies indicate that cell competition takes place between scrib clones and the surrounding wild-type tissues in the larval wing imaginal disc. Moreover, cell competition has been documented between loss-of-Dpp signal and the surrounding wild-type tissues. It is presumed that the mechanisms proposed in this study are independent of cell competition for the following reasons. First, scrib RNAi and AP-2μ RNAi data reveal that loss of BMP signal in the PCV region is produced without affecting cell polarity. Thus, cell competition is unlikely to occur in this context. Second, BMP signal is intact in scrib mutant clones of the wing imaginal disc, suggesting that cell competition caused by scrib clones is not a direct cause of loss of BMP signaling in scrib mutant cells (Gui, 2016).

Previous studies established that receptor trafficking plays crucial roles in signal transduction of conserved growth factors, including BMP signaling. Several co-factors have been identified as regulators of BMP receptor trafficking. Some of them down-regulate BMP signaling while others help maintain it. It is proposed that the Scrib-Rab5 system is a flexible module for receptor trafficking and can be utilized for optimizing a signal. During larval wing imaginal disc development, BMP ligands are trafficked through extracellular spaces to form a morphogen gradient. Although previous studies indicate that regulation of receptor trafficking impacts BMP signaling in wing imaginal discs, BMP signaling persists in scrib or dlg1 mutant cells in wing discs. Wing disc cells interpret signaling intensities of a morphogen gradient. In this developmental context, an optimizing mechanism might not be beneficial to the system. In contrast, cells in the PCV region use the system to ensure robust BMP signaling (Gui, 2016).

Taken together, these data reveal that a feedback loop through BMP and Scrib promotes Rab5 endosome-based BMP/Dpp signaling during PCV morphogenesis. Since the components (BMP signaling, the Scrib complex, and Rab5 endosomes) discussed in this work are highly conserved, similar mechanisms may be widely utilized throughout Animalia (Gui, 2016).


DEVELOPMENTAL BIOLOGY

Embryonic

To determine the subcellular localization of Scrib, antibodies were generated against Scrib peptides. In fixed tissue, Scrib is present at low levels in precellular embryos, where it is found in the actin caps that overlay the still-dividing blastoderm nuclei. As cellularization proceeds during cycle 14, Scrib staining becomes associated with the cell membranes as they invaginate basally towards the centre of the embryo. During gastrulation, however, Scrib relocates specifically to a relatively apical position along the lateral cell membrane of ectodermal cells. The apicolateral epithelial staining seen at gastrulation continues into late embryogenesis, by which time it has consolidated into a narrow subapical region (Bilder, 2000a).

Because the subapical epithelial membrane is the site of cell-cell junctions, it was determined whether Scrib localizes specifically to a junction. In mature Drosophila epithelia, the adherens junction and the septate junction (analagous to the vertebrate tight junction) are adjacent structures, located at the margins of the apical and basolateral surfaces, respectively. Co-staining for Scrib and the adherens junction marker Armadillo (Arm), the homolog of vertebrate beta-catenin, reveals that Scrib is located immediately basal to the adherens junction, whereas co-staining for Scrib and the septate junction marker Coracle (Cor), a protein 4.1 family member, reveals coincident protein distributions. These results show that Scrib localizes to septate junctions. Although the septate junction does not appear ultrastructurally until late embryogenesis at stage 14, Scrib is enriched subjacent to the adherens junction after gastrulation at stage 8. By contrast, the initial expression of Cor at stage 12 is spread throughout the basolateral membrane, with apicolateral enrichment visible only from stage 14. These data indicate that Scrib is an early marker for the site of the future septate junction, at the apical boundary of the basolateral compartment (Bilder, 2000a).

The relation between scrib, lgl, and dlg were explored by comparing the subcellular localization of the gene products. Scrib and Dlg colocalize throughout development, in particular at the apical margin of the lateral membrane (ALM) of the embryonic epidermal epithelium. Scrib is localized to the epithelial septate junction, the analog of the vertebrate tight junction, at the boundary of the apical and basolateral cell surfaces. Colocalization at the ALM occurs after gastrulation and persists in mature epithelia, where the ALM is the site of the septate junction. Lgl protein is not exclusively associated with the plasma membrane and is not polarized along it; however, it overlaps substantially with Dlg and Scrib at the ALM (Bilder, 2000b).

The tumor suppressor gene scribbled (scrib) is required for epithelial polarity and growth control in Drosophila. The identification and embryonic expression pattern is reported of two Scrib protein isoforms resulting from alternative splicing during scrib transcription. Both proteins are first ubiquitously expressed during early embryogenesis. Then, during morphogenesis each Scrib protein displays a specific pattern of expression in the central and peripheral nervous systems, CNS and PNS, respectively. During germ band extension, the expression of the longer form Scrib1 occurs predominantly in the neuroblasts derived from the neuro-ectoderm and later becomes restricted to CNS neurons as well as to the pole cells in the gonads. By contrast, the shorter form Scrib2 is strongly expressed in the PNS and a subset of CNS neurons (Li, 2001).

A P-element mutagenesis led to the identification of a recessive mutation Pvartul causing late larval lethality and producing abnormal imaginal disc growth with a complex syndrome reminiscent of that observed in mutations of tumor suppressors. Both the larval brain hemispheres and a subset of the imaginal discs displayed massive overgrowth with a loss of cell polarity. This gene was consequently classified as a tumor suppressor (Li, 2001).

The putative gene was cloned by plasmid rescue and chromosomal walk. Sequencing and characterization of isolated cDNAs and ESTs showed that it encodes three size classes of transcripts with a length of 7.2, 6.0 and 4.6 kb, respectively, sharing identical 5' ends but divergent 3' ends. Northern blot analysis confirmed the occurrence of three size classes of transcripts. Sequence alignment of the three classes of cDNAs on the genomic sequence revealed the gene structure. The 7.2 and 6.0 kb transcripts differ by the length of their 3' UTR and contain an open reading frame (ORF) of 5218 nucleotides encoding a polypeptide of 1756 amino acids that is basically identical with Scribbled and is hereafter designated as Scrib1. The alternatively spliced 4.6 kb mRNA encodes a shorter protein (Scrib2) made of 1247 residues. Both Scrib1 and Scrib2 share identical sequence over the first 1143 residues with a set of 16 leucine-rich repeats and two PDZ domains and belong to the LAP protein family. In addition, Scrib1 contains two additional PDZ domains in its unique C-terminal segment of 613 residues. Scrib2 carries a shorter C-terminal segment of 104 residues displaying no sequence similarity with the C-terminal domain of Scrib1 (Li, 2001).

To compare the expression patterns of Scrib1 and Scrib2, antibodies were raised in rabbits against a C-terminal 20-mer peptide specific for each protein. Western blots of Drosophila proteins showed that anti-Scrib1 antibodies detected a prominent band with a molecular mass of ~250 kDa, whereas the anti-Scrib2 antibodies recognized a ~180 kDa protein band. The sizes of the detected proteins correspond to those of the polypeptides translated in vitro from Scrib1 and Scrib2 cDNAs. These results show that scrib encodes two distinct polypeptides (Li, 2001).

Developmental Western blot analysis reveals that Scrib1 is expressed at a relatively high level during the first half of embryogenesis (0-12 h) and then at a lower level (12-24 h). Scrib1 expression is nearly undetectable in larvae, pupae and adults. Similarly, Scrib-2 is more intensively expressed in 0-12 h embryos than in 12-24 h embryos and remains nearly undetectable at later stages of development (Li, 2001).

Immuno-histochemistry and confocal microscopy reveals that, from the onset of blastoderm formation up to germ band elongation, Scrib1 expression is ubiquitous. Then, Scrib1 expression becomes preferentially expressed in the neuroblasts and more particularly in the developing CNS. During late embryogenesis Scrib1 is intensively expressed in a limited number of cells, including the ventral nerve chord, and is expressed at a much lower level in other epithelial structures and muscle fibers. To confirm the pattern of Scrib1 expression in the developing CNS, embryos were double stained with anti-Scrib1 and either anti-Neurotactin antibodies that recognize neuroblasts, or BP102 revealing CNS neurons. During germ band elongation Scrib1 and Neurotactin expression patterns overlap in the neuroblasts derived from the neuroectoderm. In the CNS of late embryos (stage 16-17) the most intense Scrib1 staining coincides with that of BP102 and strongly decorates the longitudinal and transversal fibers of the connectives and commissures, respectively. In addition Scrib1 is also expressed in midline cells located at the level of the anterior and posterior commissures. Strong Scrib1 expression is also detected in the pole cells of the gonads after completion of the dorsal closure (Li, 2001).

During early embryogenesis the expression of Scrib2 resembles that of Scrib1, with the difference that Scrib2 is predominantly distributed in the basal domain of the forming blastoderm cells. In contrast, Scrib1 is first localized in the apical domain and then decorates the growing plasma membrane. During late embryogenesis the tissue distribution of Scrib2 differs markedly from that of Scrib1. This is particularly evident in the CNS where Scrib2 is predominantly localized in neuronal cell bodies and Scrib1 in the axons. In addition, Scrib2 is strongly expressed in both neuronal cell bodies and projections of the peripheral nervous system (PNS). Double staining with anti-Scrib2 antibodies and 22C10 shows an intense Scrib2 expression in all 22C10 decorated cells, confirming the specific expression of Scrib2 in the PNS sensory organs (Li, 2001).

Amphiphysin family members are implicated in synaptic vesicle endocytosis, actin localization and one isoform is an autoantigen in neurological autoimmune disorder; however, there has been no genetic analysis of Amphiphysin function in higher eukaryotes. Drosophila Amphiphysin is localized to actin-rich membrane domains in many cell types, including apical epithelial membranes, the intricately folded apical rhabdomere membranes of photoreceptor neurons and the postsynaptic density of glutamatergic neuromuscular junctions. Flies that lack all Amphiphysin function are viable, lack any observable endocytic defects, but have abnormal localization of the postsynaptic proteins Discs large, Lethal giant larvae and Scribbled, altered synaptic physiology, and behavioral defects. Misexpression of Amphiphysin outside its normal membrane domain in photoreceptor neurons results in striking morphological defects. The strong misexpression phenotype coupled with the mild mutant and the lack of phenotypes suggest that Amphiphysin acts redundantly with other proteins to organize specialized membrane domains within a diverse array of cell types. In other words, Drosophila Amphiphysin functions in membrane morphogenesis, but an additional role in endocytosis cannot yet be dismissed (Zelhof, 2001).

Asymmetric cell division is important in generating cell diversity from bacteria to mammals. Drosophila neuroblasts are a useful model system for investigating asymmetric cell division because they establish distinct apical-basal cortical domains, have an asymmetric mitotic spindle aligned along the apical-basal axis, and divide unequally to produce a large apical neuroblast and a small basal daughter cell (GMC). Discs large (Dlg), Scribble (Scrib) and Lethal giant larvae (Lgl) tumour suppressor proteins regulate multiple aspects of neuroblast asymmetric cell division. Dlg/Scrib/Lgl proteins show apical cortical enrichment at prophase/metaphase, and then have a uniform cortical distribution. Mutants have defects in basal protein targeting, a reduced apical cortical domain and reduced apical spindle size. Defects in apical cell and spindle pole size result in symmetric or inverted neuroblast cell divisions. Inverted divisions correlate with the appearance of abnormally small neuroblasts and large GMCs, showing that neuroblast/GMC identity is more tightly linked to cortical determinants than cell size. It is concluded that Dlg/Scrib/Lgl are important in regulating cortical polarity, cell size asymmetry and mitotic spindle asymmetry in Drosophila neuroblasts (Albertson, 2003).


EFFECTS OF MUTATION

Because cuticles of embryos mutant for scrib suggest a broad defect in epidermal organization, the morphology of scrib embryos was examined. scrib embryos proceed normally through precellular development, and the epithelial blastoderm forms as in wild type. After gastrulation, however, organization of the ectodermal epithelium is disrupted as cells lose their columnar shape and planar arrangement. These defects become progressively more severe as development proceeds. Instead of the wild-type tightly integrated columnar monolayer epithelium, the epidermis of late scrib embryos is frequently interrupted and consists of groups of irregular rounded cells that are separating from one another. Confocal microscope sections reveal that most of the epidermis is organized into multilayered strips or tubes of cells that have lost contact with the underlying tissue (Bilder, 2000a).

The septate junction localization of Scrib suggests that cell junctions might be defective in scrib mutants. The presence of cellular junctions was investigated using antibody probes specific for either the adherens or the septate junction. The initial assembly of adherens junctions in scrib cellular blastoderms is normal, but at stage 8, anti-Arm staining reveals a severe disruption of the forming circumferential adherens junction belts. The normally continuous 'honeycomb' pattern seen in tangential apical sections of the epidermis is severely fragmented. Tissue cross-sections show that, rather than being lost, Arm is misdistributed. Instead of its wild-type localization exclusively at the apical tip of contacting cell membranes, much Arm is found around the cell periphery on the inside of the epidermis, in the midst of opposing cell membranes. At these sites, Arm colocalizes with E-cadherin (Ecad), the transmembrane component of the adherens junction, indicating that adherens junctions form at ectopic basolateral membrane positions in scrib embryos (Bilder, 2000a).

The formation of adherens junctions at ectopic positions within scrib cells raise the issue of whether these cells have lost polarity. In the wild-type epidermis, apical proteins such as Stranded at Second (Sas) are localized to the non-contacting cell membrane at the surface of the embryo, whereas basolateral proteins such as Fasciclin III (Fas III) are found in a complementary domain along the contacting membranes of cells. In scrib epidermal cells at stage 11, Sas is distributed throughout the plasma membrane, in both contacting and non-contacting cell surfaces. By contrast, Fas III localization is much less affected; in particular, it is not significantly mislocalized into apical regions. This preferential loss of apical restriction was seen with a panel of both cytoplasmic and transmembrane proteins through mid-embryogenesis, after which late embryos repolarize some apical markers by a scrib-independent pathway. Thus, the polarity defects in scrib epithelial cells arise not from a total loss of cell polarity but from a specific misdistribution of apical proteins (Bilder, 2000a).

The equivalent requirements for scrib, lgl, and dlg in epithelial development could result from independent activity of each gene in a separate pathway or from collaborative activity of the three genes in a single pathway. To address this issue, genetic interactions between the three mutations were tested and strong interactions of dlg and lgl with scrib were found. Most embryos zygotically mutant for scrib hatch and survive into late larval stages. However, embryos homozygous for scrib and additionally heterozygous for dlg die before hatching, with evident defects in dorsal closure. Dorsal closure phenotypes are characteristic of reduced activity of both dlg and lgl. Additionally, embryos homozygous for both lgl and scrib display a cuticle phenotype nearly as severe as those of lgl or scrib null embryos. Heterozygosity for lgl also enhances the imaginal disc phenotype of scrib hypomorphic larvae. These dose-sensitive interactions of dlg and lgl with scrib suggest that the three genes function in a common pathway (Bilder, 2000b).

Epistatic relations between scrib, lgl, and dlg were investigated by determining the localization of each protein in embryos mutant for the other two genes. The localization of Dlg and Scrib to the ALM was examined. In all mutant blastoderms, Scrib and Dlg are associated with ingrowing cell membranes, as in WT. However, after gastrulation, when WT embryos display an enrichment of Scrib in the ALM, lgl embryos show Scrib and Dlg localized throughout the basolateral cell membrane, a misdistribution that persists into late embryogenesis. Like lgl embryos, scrib embryos fail to polarize Dlg to the ALM, while dlg embryos not only fail to polarize Scrib to the ALM, but also display a progressive loss of membrane-associated Scrib (Bilder, 2000b).

Also examined was the distribution of Lgl, which normally has both a membrane-bound and a cytosolic component; reduction of activity in an lgl temperature-sensitive mutant correlates with loss of the membrane-bound pool. In WT embryos, Lgl is in close apposition to cell membranes. However, in scrib blastoderms and embryos, Lgl is distributed throughout the cytoplasm. dlg blastoderms shows intermediate defects in Lgl distribution, but by mid-embryogenesis loss of membrane-localized Lgl is evident. The dissociation of Lgl from the membranes of dlg embryos parallels the loss of Scrib seen in these embryos. These data indicate that dlg is required for the stable association of Scrib with the cell membrane and scrib is required for the cortical association of Lgl; all three genes act to localize Scrib and Dlg to the ALM (Bilder, 2000b).

Regulation of synaptic plasticity and synaptic vesicle dynamics by the PDZ protein Scribble

At the larval neuromuscular junction, Scribbled colocalizes and indirectly interacts with another tumor suppressor and PDZ protein, Discs-Large (Dlg). Dlg is critical for development of normal synapse structure and function, as well as for normal synaptic Scrib localization. Scrib is also an important regulator of synaptic architecture and physiology. The most notable ultrastructural defect in scrib mutants is an increase in the number of synaptic vesicles in an area of the synaptic bouton thought to contain the reserve vesicle pool. Additionally, the number of active zones is reduced in scrib mutants. Functionally, the scrib synapse behaves relatively normally at low-frequency stimulation. However, several forms of plasticity at this synapse are drastically altered in the mutants. Specifically, scrib mutants exhibit loss of facilitation and post-tetanic potentiation, and faster synaptic depression. In addition, FM1-43 imaging of recycling synaptic vesicles shows that vesicle dynamics are impaired in scrib mutants. These results identify Scrib as an essential regulator of short-term synaptic plasticity. Taken together, these results are consistent with a model in which Scrib is required to sustain synaptic vesicle concentrations at their sites of release (Roche, 2002).

Mutations in dlg lead to prominent defects in both synapse structure and function. At the ultrastructural level these defects include an increase in bouton size and number of active zones, as well as a poorly developed subsynaptic reticulum (SSR), an elaborate folding of the postsynaptic membrane at the NMJ. Dlg is colocalized with Scrib, and mutations in dlg also result in severe mislocalization of synaptic Scrib. In contrast, although localization of Scrib to the NMJ is completely disrupted in scrib mutants, the localization of Dlg is not affected. Analysis of the NMJs in scrib mutants has shown that the general morphology is not affected. It is hypothesized that some of the defects in dlg mutants might be the consequence of Scrib mislocalization. This hypothesis was tested by serially sectioning type I synaptic boutons in several scrib mutant allelic combinations and examining their ultrastructure using electron microscopy. It was found that the synaptic structure in these mutants was drastically altered; however, these defects were quite distinct from those in dlg mutants. One of the most prominent defects was an abnormally high density of synaptic vesicles. In wild type, synaptic vesicles are organized into at least two pools: a pool in direct proximity to the T-shaped active zones [thought to represent the readily releasable pool (RRP)], and a pool localized in a broad area at the periphery of the entire synaptic bouton [representing the reserve vesicle pool (RP)]. Typically, the central region of the bouton is devoid of synaptic vesicles and contains endosomes and mitochondria, as well as other nonvesicular material. In contrast, boutons in the null allele scrib2 and scrib2/Df, but not those from the less severe allele scrib1, are filled with synaptic vesicles and lack an empty core. The number and area of mitochrondrial profiles, however, is unchanged in scrib mutant boutons. Overall, in these mutants there is a significant increase in synaptic vesicle density, as measured by determining the total number of vesicles at the central cross section of the boutons divided by the area of this cross section. In addition to this striking defect in vesicle distribution and density, many boutons contained morphologically abnormal vesicular material at the core (Roche, 2002).

To determine whether both the RRP and the RP are affected in scrib mutants, the number of vesicles was counted in an area 100, 150, and 200 nm around the active zone, which likely encompasses the RRP. The number of vesicles in these areas of scrib2/Df mutant boutons was not significantly different from wild type. Thus, it is the distribution and density of the RP that appear to be specifically affected in scrib mutants (Roche, 2002).

In addition to the defect in the RP, the average number of active zones in both scrib2 and scrib2/Df is slightly lower than wild type, although this difference was statistically different only at scrib2 homozygous boutons. Unlike dlg mutants, the SSR appeared normal, and neither the number of SSR layers nor the SSR density is significantly different from wild-type controls. This is in contrast to the observations in severe dlg mutants, in which the number of active zones is increased several fold and the SSR length is reduced (Roche, 2002).

Another phenotype in scrib mutants is the presence of an abnormally thick basal lamina. In ~30% of boutons examined, the muscle basal lamina appeared to be composed of several layers of normally sized basal lamina. However, at the majority of scrib mutant boutons, the basal lamina had a uniform electron density. These data suggest that scrib is involved in the normal development of several aspects of synapse structure (Roche, 2002).

Transgenic Scrib was expressed in motor neurons and muscles of scrib2 mutants, using the Gal4 drivers C380 and BG487. In these transgenic animals the density of synaptic vesicles was significantly decreased from the levels found in scrib mutants. In fact, the density of synaptic vesicles was significantly lower even than the densities in wild-type boutons. A similar effect was seen when analyzing the number of active zones, which was significantly higher than not only scrib mutants but also wild-type boutons. These data suggest that expression of Scrib in the mutant background not only rescues the scrib mutant defect, but causes a new phenotype in the opposing direction and likely results from an overexpression of Scrib compared with wild type. In contrast, the defect in the basal lamina was only partially rescued by transgenic Scrib expression. Thus, both the synaptic vesicle density and the number of active zones can be modified in either direction by manipulating the concentration of Scrib at the synapse (Roche, 2002).

To understand the physiological significance of the structural defects observed in scrib mutants, synaptic function was examined. Both evoked synaptic events [excitatory junctional currents (EJCs)] and spontaneous events [miniature EJCs (mEJCs)] were measured by two-electrode voltage-clamp experiments in muscle 6 of third instar larvae. Changes in the frequency of miniature synaptic currents result from alterations in presynaptic function, caused by alterations either in the probability of release (pR) or in the number of release sites. Conversely, changes in the amplitude of miniature events are generally considered to result from postsynaptic alterations, usually a change in receptor concentration or function. In scrib2/Df larvae, there was a significant decrease in the frequency of miniature synaptic events. Additionally, there was a slight but significant decrease in the amplitude of miniature synaptic events. Notably, presynaptic expression of transgenic Scrib using the Gal4 driver C380 in the scrib2 mutant background not only rescued the decrease in mEJC frequency but resulted in a significant increase in mEJC frequency compared with wild-type controls. This is in striking agreement with the ultrastructural studies, which show that expression of transgenic Scrib not only rescues the decrease in active zone number but results in a significant increase in the number of active zones compared with wild type. This result also is a strong indicator that, in this case, mEJC frequency is a reflection of the number of release sites (Roche, 2002).

Presynaptic expression of Scrib in the scrib2/Df background has no effect on mEJC amplitude, indicating that the reduced mEJC amplitude in scrib2/Df larvae arises from a postsynaptic mechanism (Roche, 2002).

Evoked release was measured by stimulating the segmental nerve innervating muscle 6 using a glass suction electrode. The nerve was stimulated with suprathreshold voltage at a frequency of 1 Hz, and the resulting EJCs were recorded. Surprisingly, there was no significant change in the amplitude of evoked EJCs when scrib2/Df was compared with wild type. There was a small but statistically insignificant increase in the quantal content of scrib2/Df larvae resulting from the small decrease in mEJC size. There was a slight divergence in wild-type and scrib mutant EJC amplitudes at low Ca2+ concentrations, with a significant difference only at 0.3 mM Ca2+. A double logarithmic plot of EJC amplitude in the linear portion of the Ca2+ concentration curve, an indicator of Ca2+ cooperativity, reveals a slight change in the slope of the linear fit, from 3.4 ± 0.1 for wild type to 2.8 ± 0.4 for scrib2/Df. Thus, vesicle release in scrib2/Df is slightly more sensitive to external Ca2+ levels than wild-type larvae. However, this change in sensitivity is very small in comparison to other Drosophila mutations that alter the Ca2+ sensitivity of vesicle release, perhaps a result of a mislocalization of release machinery with respect to cytoplasmic Ca2+ rather than a complete elimination of a critical component of the Ca2+ sensing process (Roche, 2002).

The similarity of evoked responses was surprising in view of the drastic changes in the ultrastructure and the decrease in spontaneous release frequency at synapses of the scrib mutant larvae. The response to high-frequency stimulation was tested to determine whether underlying defects may be uncovered during conditions in which the speed and accuracy of vesicle dynamics play a more crucial role. In wild type at 0.5 mM Ca2+, 10 Hz stimulation results in short-term facilitation of the synaptic current. This is thought to be caused by increased vesicular release resulting from residual Ca2+ remaining in the neuronal cytoplasm from previous depolarizations, acting on unknown targets to increase vesicle release. In contrast to wild type, facilitation was severely reduced or absent in scrib2/Df mutants, and unlike some other mutants that do not exhibit facilitation, the baseline transmission of scrib2/Df was equivalent to wild type at 0.5 mM Ca2+. Presynaptic expression of transgenic Scrib restored the response of the synapse to high-frequency stimulation (Roche, 2002).

To test whether the effect seen at 0.5 mM Ca2+ resulted from the specific inability to show facilitation or was a more global defect in synaptic vesicle replenishment at high stimulation frequencies, the synaptic response was tested at higher Ca2+ concentrations (1.0 mM), where no facilitation is seen in wild-type larvae. At 1.0 mM Ca2+, little change in EJC amplitude is seen after switching to 10 Hz stimulation frequencies, presumably because of a balance between the number of vesicles released and the resupply of the RRP. In contrast to the maintained EJC amplitude seen in wild-type larvae at 10 Hz, a significant depression is seen in scrib2/Df mutants, becoming apparent immediately after the first few stimuli. Thus, high-frequency stimulation causes scrib2/Df mutant synapses to exhibit both a lack of facilitation at low Ca2+ concentrations and faster synaptic depression at higher Ca2+ concentrations. These data suggest that scrib2/Df is defective not in the ability to exhibit facilitation but rather in the ability to resupply the RRP during high-frequency firing (Roche, 2002).

Another form of activity-dependent short-term plasticity exhibited at the Drosophila NMJ is post-tetanic potentiation (PTP). After a short train (30 sec) of high-frequency (10 Hz) stimuli, the EJC is potentiated for a period of 3-4 min relative to EJCs before the tetanus. The amplitude of wild-type EJCs is potentiated more than twofold after the train of high-frequency stimulation. scrib2/Df, in contrast, shows only a modest potentiation after the same stimulation protocol. Thus, multiple forms of activity-dependent plasticity are altered at the scrib synapse (Roche, 2002).

The alteration in short-term plasticity observed in response to high-frequency stimulation in scrib mutants, combined with the ultrastructural observations showing a remarkable increase in vesicle density, suggest that vesicle dynamics might be affected in the mutants. For example, if the rapid recruitment of vesicles to the RRP is altered, then synaptic transmission is expected to be normal at low stimulation frequencies, because at these frequencies the RP does not contribute to vesicle release. However, at high frequencies the RRP size must be maintained by rapid recruitment of vesicles. The vesicles are replenished to some degree by endocytosis of recently released vesicles. The increased density of vesicles in scrib mutants suggests that the defects in short-term plasticity are not likely a result of vesicle depletion, as is seen in the endocytotic mutant shibire, but more likely a result of the inability to rapidly recruit vesicles into the RRP. This hypothesis was tested by activity-dependent labeling of recycling vesicles using the styryl dye FM1-43 (Roche, 2002).

For these studies, preparations were dissected in 0.1 mM Ca2+ HL-3 saline and subsequently depolarized for 2 min in HL-3 saline containing 90 mM K+ in the presence of 5 µM FM1-43. After washing and fixing, samples were imaged by confocal microscopy. In wild type, strong FM1-43 fluorescence was observed at type I boutons. This labeling paradigm has been shown to label what has been termed the exo/endo cycling pool (ECP) in Drosophila, a pool that has been shown to contribute to both low- and high-frequency vesicle release (Roche, 2002).

This loading procedure resulted in fluorescence that was observed in a broad area at the periphery of synaptic boutons, whereas the central core of the boutons was devoid of fluorescence. This is in agreement with reports showing a similar distribution of synaptic vesicles. In scrib2/Df mutants a 54 ± 5% reduction in FM1-43 fluorescence was found compared with wild-type levels. In addition, the distribution of the FM1-43 label was affected in the mutants, which showed a significantly larger percentage of boutons with diffuse FM1-43 throughout the bouton surface. Thus, scrib2/Df mutants exhibit a decrease in activity-dependent endocytosis or altered distribution of endocytosed vesicles, or both. Because scrib2/Df larvae have very high synaptic vesicle densities in contrast to endocytotic mutants such as shibire and AP2, which show synaptic vesicle depletion, it is proposed that the defect in scrib mutants arises from a defect in exocytosis that secondarily causes effects on vesicle distribution, rather than a specific defect in endocytosis or endocytotic sorting. Indeed, the number of vesicles contained in the ECP is known to affect further loading of synaptic vesicles (Roche, 2002).

These data indicate that Scrib has a prominent role in synapse function and, taking into account its multiple PDZ domains, is likely involved in the precise localization of proteins necessary for vesicle dynamics. Facilitation is a Ca2+-dependent process that takes place in many synapses. This process is thought to result from residual Ca2+ accumulation, which acts on unidentified components of the exocytotic pathway. The precise mechanism by which high-frequency stimulation may lead to an increase in quantal content is currently unknown; however, several possibilities exist, including an increase in the pR of docked vesicles or an increase in the number of docked vesicles. There are several mutations in Drosophila that have effects on facilitation and PTP, including dunce (cAMP-specific phosphodiesterase II), rutabaga (Ca2+/calmodulin-dependent adenylyl cyclase), volado (alphaPS3-integrin), leonardo (a 14-3-3 protein family member), and latheo (an origin of replication complex protein). Of these mutants, scrib behaves most like rutabaga and latheo, in that both of these lack facilitation when baseline synaptic transmission is equivalent to wild type, and both have altered Ca2+ sensitivity of vesicle release, although the alterations in Ca2+ sensitivity are much more dramatic in both rutabaga and dunce mutants than in scrib mutants. It will be interesting to determine whether the localization of either of these proteins is altered in scrib mutants (Roche, 2002).

Three alternative models, or a combination of these, may contribute to the functional abnormalities observed in scrib mutants. One possibility is that the lack of facilitation observed at low Ca2+ in scrib mutants is caused by the inability of these synapses to recruit, transport, or convert vesicles into the RRP. This notion is supported by the FM1-43 studies, showing that endocytosis of vesicles in scrib mutants is greatly reduced, because endocytosis would be influenced by the amount of vesicles released as well as the number of vesicles already contained in the synapse. In other words, the cellular machinery underlying facilitation may be intact in scrib mutants, but the inability to recruit additional vesicles may mask the expression of facilitation. Similarly, during high-frequency stimulation at higher Ca2+, a decreased capability to recruit vesicles to the RRP would result in increased magnitude of depression in scrib mutants (Roche, 2002).

A second model is based on evidence indicating the existence of a feedback mechanism at the Drosophila NMJ that operates to maintain the excitability of the muscle within a narrow range. Because of this compensatory mechanism, the decrease in number of active zones in scrib mutants may lead to a maximal increase in pR, thus maintaining EJC amplitude at low frequencies. Stimulation of the terminal at frequencies that in wild type lead to facilitation, however, would be unable to further increase pR in scrib mutants, precluding further increases in the amplitude of the EJC. In other words, compensatory mechanisms in scrib mutants bring the mutant synapse to a facilitated state even at low-frequency stimulation, preventing a further increase in EJC amplitude at higher frequencies. The rapid depression observed in scrib mutants could be explained according to this model by the inability of the terminal to fulfill the increased demand for primed vesicles that is imposed by the increase in pR. In intact Drosophila larvae, motoneurons stimulate the muscles by firing trains of high-frequency action potentials. In scrib mutants, loss of high-frequency responsiveness might lead to the buildup of synaptic vesicles, perhaps explaining the enhanced density of synaptic vesicle in scrib mutants. Thus this model is consistent with the electrophysiological and ultrastructural observations in the mutants (Roche, 2002).

A third possible mechanism involves altered buffering of neuronal Ca2+ levels. Cytoplasmic Ca2+ buffering is important in regulating synaptic vesicle release, especially during high-frequency stimulation. A prevalent form of buffering Ca2+ at synapses is mitochondrial Ca2+ uptake, and this form of Ca2+ uptake has been shown to have effects on high-frequency synaptic firing patterns. The number of mitochondria in scrib boutons, however, was unchanged from wild type, making this possibility less likely. However, elimination or mislocalization of other critical Ca2+ buffering or sensing molecules may play a role in the defects seen in scrib synapses. Ultimately, the identification of Scrib binding partners may shed light on the precise mechanisms by which facilitation and sustained release are affected in scrib mutants (Roche, 2002).

Another defect in scrib mutant synapses was an abnormally thick basal lamina. The synaptic basal lamina has long been recognized as containing important elements for postsynaptic differentiation and for the clustering of neurotransmitter receptors. The effects of Scrib on the basal lamina may reflect the inability of Scrib to selectively recruit synaptic components to their correct destination (Roche, 2002).

PDZ proteins are characterized by multiple modular sequences involved in protein-protein interactions and for this reason are frequently called scaffolding proteins. These proteins bring together components necessary for certain cellular functions by binding to distinct partners through their multiple interaction domains. Scrib contains four PDZ domains and hence could form a multiprotein complex with at least four different proteins. Elucidation of the identity of these binding partners will be important in understanding the precise role that Scrib plays in synaptic vesicle dynamics. In epithelial cells it has been suggested that Scrib is involved in vesicle sorting, an important mechanism involved in segregating transmembrane proteins in these cells. Another PDZ domain-containing protein, EBP50, has also been shown to be involved in vesicular sorting, in particular, sorting of endocytotic vesicles involved in recycling ß adrenergic receptors. A similar mechanism, perhaps operating through a conserved cassette of proteins, may be altering vesicle dynamics at the synapse (Roche, 2002).

One might hypothesize, on the basis of the mislocalization of Scrib in dlg mutants, that by mutating dlg one would see, in addition to a dlg-specific phenotype, the scrib phenotype as well. Indeed this seems to be the case in other cell types in which mutation of either dlg or scrib causes similar phenotypes: formation of tumors and loss of cell polarity. However, at the Drosophila NMJ the effects of scrib mutation are quite different from those in several dlg mutants. These differences may stem from the fact that residual synaptic Scrib is still present in dlgXI-2 mutants, although at a lower level, and hence the remaining Scrib may be sufficient to override the synaptic scrib phenotype. Interestingly, levels of synaptic Scrib have an opposite influence on the regulation of the number of active zones than do levels of synaptic Dlg. Although a decrease in Dlg levels in severe dlg mutants causes an increase in active zone number, the same phenotype is observed by increasing Scrib levels. This observation is consistent with the notion that at synapses Scrib may negatively regulate Dlg function. This is in contrast to the observation in epithelial cells, where Dlg and Scrib appear to function in a similar manner during the determination of cell polarity and tumor suppression. This may reflect the ability of Dlg and Scrib to bind different protein partners with different functions in the two cell types. Indeed, partners such as Fasciclin II bind to Dlg at synapses but are absent in epithelial cells. Thus, the specific influence of scaffolding proteins in different cell types may be highly dependent on the availability of specific binding partners (Roche, 2002).

In conclusion, this study has introduced a relatively new function for a member of the PDZ protein family, regulation of synaptic vesicles. It will be important to understand the specific subset of proteins that interacts with Scrib, because this is likely the key to understanding how this protein is involved in synaptic vesicle regulation (Roche, 2002).

scribbled mutants cooperate with oncogenic Ras or Notch to cause neoplastic overgrowth in Drosophila

Cancer is a multistep process involving cooperation between oncogenic or tumor suppressor mutations and interactions between the tumor and surrounding normal tissue. This study is the first description of cooperative tumorigenesis in Drosophila, and uses a system that mimics the development of tumors in mammals. The MARCM system was used to generate mutant clones of the apical-basal cell polarity tumor suppressor gene, scribbled, in the context of normal tissue. scribbled mutant clones in the eye disc exhibit ectopic expression of cyclin E and ectopic cell cycles, but do not overgrow due to increased cell death mediated by the JNK pathway and the surrounding wild-type tissue. In contrast, when oncogenic Ras or Notch is expressed within the scribbled mutant clones, cell death is prevented and neoplastic tumors develop. This demonstrates that, in Drosophila, activated alleles of Ras and Notch can act as cooperating oncogenes in the development of epithelial tumors, and highlights the importance of epithelial polarity regulators in restraining oncogenes and preventing tumor formation (Brumby, 2003).

A clonal approach, more closely resembling the clonal nature of mammalian cancer, was used to analyze the effects of removing Scrib function on tumor formation. This analysis indicates that Drosophila scrib- tumors: (1) lose tissue architecture, including apical-basal cell polarity; (2) fail to differentiate properly; (3) exert non-cell-autonomous effects upon the surrounding wild-type tissue; (4) upregulate cyclin E and undergo excessive cell proliferation; (5) are restrained from overgrowing by the surrounding wild-type tissue via a JNK-dependent apoptotic response, and (6) show strong cooperation with oncogenic alleles of Ras and Notch to produce large amorphous tumors. These conclusions are summarized in a model for tumor development in Drosophila. It is suggested that the role of epithelial cell polarity regulators in restraining oncogenes is likely to be of general significance in mammalian tumorigenesis (Brumby, 2003).

The model suggests that the wild-type larval eye disc is a monolayered columnar epithelium, in which cell proliferation is tightly regulated. Cell architecture is maintained by the formation of adherens junctions, the apical localization of Scribbled, and adhesion to the basement membrane. Mutation of scrib results in loss of apical-basal polarity, leading to multilayering and rounding up of cells. scrib- tissue also shows impaired differentiation, and ectopic cyclin E expression (by an unknown mechanism) leads to ectopic cell proliferation. Unrestrained overgrowth and tumor formation of scrib- cells is held in check by compensatory JNK-mediated apoptosis, dependent upon the presence of surrounding wild-type cells. Secondary mutations are required to avoid this apoptotic fate. If JNK activity is blocked within scrib- cells, by expressing a dominant-negative form of JNK, apoptosis is prevented, resulting in tissue overgrowth and lethality. Even more aggressive overgrowth results from the addition of activating oncogenic alleles of Ras or Notch. In addition to promoting cell survival, these oncogenes must also promote tumor cell proliferation; however, it is proposed that other downstream effectors of these oncogenes are likely also to be important, since it was not possible to mimic the cooperative overgrowth effects of RasACT or NACT on scrib- tissue by simply blocking apoptosis and enhancing cell proliferation (Brumby, 2003).

scrib- clones ectopically express cyclin E and undergo ectopic S phases and mitoses. Since cyclin E is rate limiting for cell cycle progression in the developing eye, it is likely that upregulation of cyclin E in scrib- clones is critical for the ectopic cell proliferation. Indeed, alleles of scrib and lgl were originally isolated as dominant suppressors of a hypomorphic cyclin E allele, DmcycEJP, suggesting that these cell polarity genes normally play a critical role in limiting cyclin E expression. Currently being investigated is which signaling pathways are altered in scrib, dlg or lgl mutants that could be responsible for cyclin E upregulation. A recent study in human lung epithelial cells shows that disrupting cell polarity allows mixing of the heregulin-alpha ligand and the erbB2-4 receptor, which are normally physically separated, resulting in activation of the pathway and cell proliferation. Further studies are required to determine whether the ectopic expression of cyclin E observed in the absence of Scrib is simply a consequence of the tissue disorganization induced by disrupting cell polarity, or if Scrib has a direct role in limiting cell proliferation independent of cell polarity. Interestingly, the rounding up of cells in the absence of Scrib appears to be predominantly a cell-autonomous effect, yet clearly non-cell-autonomous defects are also apparent, including the upregulation of cyclin E. This would suggest that altered cell-cell interactions between wild-type and mutant cells can also alter signaling pathways within wild-type cells, and that the loss of apical-basal polarity and collapse of the columnar epithelium is not intrinsically responsible for the deregulated expression of cyclin E. A deeper understanding of the relationship between epithelial cell polarization and cell proliferation is clearly important for understanding the development of cancer, since a loss of cell polarity often accompanies tumor progression and metastasis (Brumby, 2003).

Overproliferation of scrib- clones in the eye disc is compensated for by JNK-mediated apoptosis. Blocking JNK pathway activity in scrib- eye clones greatly increases the proportion of clonal tissue, and results in lethality to the host. Since downregulating JNK pathway activity in otherwise wild-type clones of tissue does not induce increased cell proliferation, it is suggested that JNK pathway activity in scrib- clones induces apoptosis. This is consistent with previous reports on the pro-apoptotic effects of the JNK pathway in the Drosophila eye and the current observations. Recent studies in mammals would also suggest that activation of the JNK pathway can limit the growth of tumors in situ, possibly by increasing apoptosis (Brumby, 2003).

How JNK-mediated apoptosis is induced in scrib- clones is not known. While Scrib could play a direct role in repressing JNK pathway activity, it is also possible that the JNK pathway is activated indirectly, in response to other cellular defects. In the wing disc, removal of cells by JNK-mediated apoptosis is linked to discontinuities in a cell's response to morphogen gradients, most notably the antero-posterior patterning regulator, Dpp, in a process probably related to cell competition, with the purpose of eliminating aberrant or slow growing cells. Although this form of compensatory JNK-mediated apoptosis has not yet been demonstrated within the eye disc, the observation that the surrounding wild-type tissue context plays an important role in limiting the overgrowth of scrib- tissue argues against a simple cell-intrinsic apoptotic response of scrib- cells to a loss of cell polarity, and is more consistent with an integrative response mediated by both the tumor cells and the surrounding wild-type cells, as exemplified by cell competition. Whether this is dependent on a failure of scrib- cells to transduce Dpp signaling is not known; however, other interesting possibilities also warrant further investigation. Notable is the recent identification of a tumor necrosis factor-induced apoptotic signaling pathway involving the JNK pathway. It is also important to keep in mind the involvement of the JNK pathway in orchestrating cell shape changes during the morphogenetic movements of dorsal and thorax closure and wound healing. clones of scrib- tissue expressing BskDN (JNKDN) appear morphologically different from those expressing the apoptosis inhibitor p35; most notably, the clones are generally larger and less rounded than those expressing p35. This would imply either that p35 is not as effective as BskDN in preventing cell death, or that there are other effectors of the JNK pathway that are important in the inhibition of scrib- tumor overgrowth. The possibility that JNK activation could play a role in eliminating scrib- tissue from the epithelium in a process reminiscent of wound healing is currently being investigated (Brumby, 2003).

dlg and lgl mutant clones also show poor viability, suggesting that JNK-mediated apoptosis could be a common response to the loss of cell polarity and overproliferation induced by the absence of these tumor suppressors. Indeed, while other regulators of epithelial cell polarity, such as Crumbs and E-cadherin, apparently do not act as tumor suppressors in Drosophila, the effects of these mutations on cell proliferation when cell death is blocked warrant further examination. Interestingly, in mammalian systems, the polarized nature of epithelia is also important in protecting cells from an apoptotic response, and this acts as a brake on tumor development when polarity is disrupted (Brumby, 2003).

In Drosophila, activated Ras exerts its oncogenic effects through Raf and the MAPK pathway. Downstream targets of MAPK in the eye disc promote differentiation, cell survival and cell proliferation. This work also demonstrates that Ras can increase cyclin E protein levels in the eye disc. In combination with scrib-, the differentiation output of RasACT signaling appears to be attenuated, and the proliferative and anti-apoptotic responses prevail (Brumby, 2003).

Activated Notch also cooperates with scrib-, resulting in neoplastic overgrowth, and although no anti-apoptotic role for Notch signaling in the eye has been described previously, NACT exerts hyperproliferative effects in flies, and Notch signaling is required for proliferation of eye disc cells. Although it is not known if NACT induces the same critical downstream targets as RasACT to cause overgrowth of scrib- tissue, removing ras function in scrib- cells overexpressing NACT rescues the overgrowth phenotype, suggesting that the effects of NACT are at least partially dependent on Ras (Brumby, 2003).

Initially it seemed likely that the cooperative effects of RasACT or NACT on scrib- tissue could be explained by the ability of these oncogenes to promote cell proliferation while blocking apoptosis. However, the expression of neither cyclin E nor E2F1/DP, in combination with the apoptosis inhibitor p35 (or with the inhibitor of JNK pathway activity, BskDN), was capable of phenocopying the effect of RasACT or NACT in scrib- clones. It is therefore suggested that other downstream effectors, apart from anti-apoptotic and cell cycle regulators, must be important in mediating the oncogenic effects of RasACT or NACT. In fact, in Drosophila, Ras has also been shown to be a potent inducer of cellular growth, while cyclin E and E2F1 mainly promote cell cycle progression. Whether NACT also promotes cell growth in Drosophila has not been examined in detail. If growth promotion targets downstream of RasACT or NACT are critical in promoting the overgrowth of scrib- tumors, these are likely to be independent of the PI3 kinase pathway since ectopic PI3 kinase signaling in scrib- clones does not induce synergistic overgrowth, and RafACT is able to induce overgrowth as equally extensive as RasACT (Brumby, 2003).

Finally, it is noted that in mammalian systems, evidence exists for a role for Ras signaling in modulating cell junction complexes and enhancing epithelial to mesenchymal transitions, and in Drosophila also, constitutive RasACT signaling in clones alters cell affinities and changes the levels of E-cadherin and ß-catenin. Whether RasACT or NACT signaling destabilizes adherens junctions in Drosophila and this potentiates scrib- neoplastic overgrowth or whether alterations in the structure of the adherens junction resulting from the absence of Scrib alters a cells response to constitutive activation of these oncogenes are important future questions (Brumby, 2003).

This study has described a novel multi-hit model of tumorigenesis in Drosophila. Furthermore, although it has been suspected that disruptions to cell polarity could potentiate tumor progression and metastasis, this work demonstrates for the first time how the oncogenic effects of activated Ras and Notch are unleashed in the absence of epithelial polarity regulators. It is predicted that in mammals also, defects in apical-basal polarity could cooperate with oncogenes during neoplastic development. This approach in Drosophila can now be used to screen for novel oncogenes that, when specifically overexpressed in scrib- clones, are capable of inducing cooperative tumorigenesis, and can also be extended to identify cooperative interactions between other tumor suppressors and oncogenes within a whole animal context (Brumby, 2003).

Oncogenic mutations produce similar phenotypes in Drosophila tissues of diverse origins

An emerging interest in oncology is to tailor treatment to particular cancer genotypes, i.e. oncogenic mutations present in the tumor, and not the tissue of cancer incidence. Integral to such a practice is the idea that the same oncogenic mutation(s) produces similar outcomes in different tissues. To test this idea experimentally, tumors were studied driven by a combination of RasV12 and scrib1 mutations in Drosophila larvae. Tumors induced in tissues of neural ectodermal and mesodermal origins were found to behave similarly in every manner examined: cell cycle checkpoints, apoptosis, cellular morphology, increased aneuploidy and response to Taxol. It is concluded that oncogenic effects override tissue-specific differences, at least for the mutations, tissues, and phenotypes in this study (Stickel, 2014).

Scribble is essential for olfactory behavior in Drosophila melanogaster

The ability to discriminate and respond to chemical signals from the environment is an almost universal prerequisite for survival. Reported here is evidence that the scaffold protein Scribble is essential for odor-guided behavior in Drosophila. A P-element insert line has been identified with generalized sexually dimorphic smell impairment, smi97B. The transposon in this line is located between the predicted promoter region and the transcription initiation site of scrib. A deficiency in this region, Df(3R)Tl-X, and two scrib null alleles fail to complement the smell-impaired phenotype of smi97B. Wild-type behavior is restored by precise excision of the P element, scrib mRNA levels correspond with mutant and wild-type phenotypes, and introduction of a full-length scrib transgene in the smi97B mutant rescues the olfactory deficit. Expression of Scrib is widespread in olfactory organs and the central nervous system. Finally, alternative splicing of scrib generates transcripts that differ in the number of leucine-rich repeats and PDZ domains (Ganguly, 2003).

The smi97B mutation is one of the strongest smi mutations among a set of previously identified smi lines. The mutation is recessive: olfactory ability, quantified by avoidance responses to benzaldehyde, was reduced in smi97B homozygotes, compared to the P-element-free coisogenic host strain, Sam; ry506 (Sam), whereas smi97B/Sam heterozygotes display avoidance responses that are indistinguishable from wild type. Furthermore, the magnitude of the mutational effect of smi97B is sexually dimorphic; even at high odorant concentrations: males are hyposmic, while females are anosmic (Ganguly, 2003).

To determine whether smi97B flies experience smell impairment throughout their life cycle, whether larvae also display aberrant olfactory responsiveness was examined. Since larvae are attracted to most odorants, even those that elicit avoidance behavior in adults, the kinetics were compared of odor-guided responses of wild-type and smi97B third instar feeding larvae toward benzaldehyde and isoamylacetate. The effect of genotype in the two-way analysis of variance was significant for both odorants, but time and genotype x time interaction terms were not significant for either odorant. Larval motility in the absence of an odor cue was not significantly different between the two genotypes. Thus, the smi97B mutation causes olfactory deficits in both larvae and adults (Ganguly, 2003).

The genomic fragment flanking the 3' end of the P[lArB] element was sequenced and the insertion site was determined to be 1084 bp upstream of the open reading frame of scrib. scrib is a pleiotropic gene essential for localization of polarity determinants in developing epithelia, and synaptic maturation and modulation of short-term plasticity at the larval neuromuscular junction. smi97B is an allele of scrib (Ganguly, 2003).

The smi97B mutation was mapped to the region of the third chromosome including scrib. Df(3R)Tl-X (breakpoints 97B2;97D2), which uncovers scrib, failed to complement smi97B; hemizygotes generated by crossing Df(3R)Tl-X to smi97B displayed reduced avoidance responses compared to Df(3R)Tl-X/Sam controls. The smell-impaired phenotype of Df(3R)Tl-X/smi97B hemizygotes was also sex specific, with significantly more smell impairment in females than in males (Ganguly, 2003).

Next, to demonstrate that olfactory deficits in smi97B arise from the P[lArB] element and are not a linked mutation, precise excision of P[lArB] was shown to restore the wild-type phenotype; avoidance responses of precise excision alleles, like smi97B16A, were not significantly different from Sam. Further, mutations generated by imprecise excision of P[lArB] provided evidence for sex-specific regulation of scrib. The smi97B15A mutation contains a 3.6-kb P[lArB] fragment at the original insertion site that resulted in male-specific olfactory deficits: smi97B15A males were smell impaired compared to controls, while olfactory responses in females were not statistically different from those in wild type. In contrast, a 2.5-kb P[lArB] insertion at the same site in smi97B2A was correlated with mild hyposmia in females. In agreement with the sexually dimorphic phenotype, scrib transcripts were markedly reduced in smi97B15A males, but not in females, whereas transcriptional differences could not be resolved in smi97B2A, in line with the subtle female-specific phenotype of this imprecise revertant (Ganguly, 2003).

To further implicate scrib in olfactory behavior, complementation tests were conducted with previously identified scrib alleles. Two null alleles, scrib1 and scrib2, failed to complement the smell-impaired phenotype of smi97B. Avoidance responses of scrib1/smi97B and scrib2/smi97B heterozygotes were significantly lower than those of the scrib1/Sam and scrib2/Sam controls. No sexual dimorphism in smell impairment was observed, possibly due to the disparate genetic backgrounds of the scrib stocks and smi97B. scribS0421405 and scribj7B3 alleles, which contain P[lacW] insertions in the 5' untranslated region of scrib and the second intron, respectively, were tested. Avoidance responses of scribS0421405/smi97B females were significantly lower than those of scribS0421405/Sam females, while male responses were not significantly different from those of control males. Hence, scribS0421405 failed to complement the olfactory deficit caused by smi97B, but only in females. However, scribj7B3 fully complemented the smell-impaired phenotype of smi97B; avoidance responses of scribj7B3/smi97B flies were not significantly different from those of scribj7B3/Sam controls. Interallelic complementation is consistent with alternative splicing of scrib, which may involve the generation of sex-specific gene products involved in olfaction. Rescue of the smi97B phenotype was demonstrated by functional complementation with a wild-type scrib allele (Ganguly, 2003).

The sexually dimorphic olfactory phenotype of smi97B and evidence for interallelic complementation led to an examination of whether males and females express alternative splice variants of scrib. Three major RNA species were detected on Northern blots probed with a full-length scrib cDNA: a universal 5.9-kb transcript present in adults and to a lesser extent in larvae, a 7.1-kb transcript expressed predominantly in males and larvae of both sexes, and a 4.6-kb female-specific transcript. Transcript levels were correlated with sex-specific behavioral phenotypes, as evidenced by reductions in the 7.1-kb transcript in smi97B larvae and the 5.9- and 4.6-kb transcripts in mutant females compared to controls (Ganguly, 2003).

To investigate the existence of less prominent variants of scrib, a Drosophila head cDNA library was screened with a full-length scrib probe. Seven unique clones were identified and sequenced. The sizes of three inserts correspond with splice variants detected on Northern blots. A 7.1-kb clone encoding 16 LRRs, four PDZ domains, and a unique 3' untranslated exon corresponds to the transcript expressed in males and larvae; a 5.9-kb clone identical to the 7.1-kb fragment, but without the 3' untranslated region, corresponds to the transcript present in both sexes and larvae; and a 4.6-kb clone encoding 16 LRRs and PDZ domains I and II corresponds to the female-specific transcript. In addition, the 4.6-kb clone also contains a unique coding exon at the 3' end. The first 285 bases of this exon were shared by four additional clones, two of which were identical except for the number of LRRs they encode. In contrast to the 4.6-kb female-specific transcript that encodes only PDZ domains I and II, two clones were identified that encode only PDZ domains III and IV. Since LRRs and PDZ domains mediate protein-protein interactions, these differences suggest variability in the composition of protein assemblies recruited by the various Scrib isoforms (Ganguly, 2003).

Monospecific antibodies raised against a carboxyl-terminal peptide of Scrib did not visualize the expected 200-kD polypeptide encoded by the transgene. Instead they detected a 120-kD band in both sexes and an 80-kD female-specific immunoreactive band, likely due to high sensitivity of the protein in adult flies to proteolysis, which cleaves the expected 200-kD translation product in 120- and 80-kD immunoreactive polypeptides in females, whereas in males the latter fragment is proteolyzed further into smaller fragments (Ganguly, 2003).

Visualization of scrib expression in adult tissues with a riboprobe complementary to the scrib coding region revealed staining in the third antennal segment and maxillary palps and the major olfactory organs of Drosophila as well as in Johnston's organ in the second antennal segment, the primary auditory organ. Staining was also observed in cortical regions of the brain. Staining was not observed when hybridizations were performed with sense riboprobes. No differences in scrib expression were observed between males and females under these experimental conditions (Ganguly, 2003).

To localize Scrib in CNS projection areas, immunohistochemistry was performed. Staining in the brain was particularly intense in the antennal nerves and the ventrolateral and superior medial protocerebrum. Widespread deposition of Scrib was also detected in the antennae and maxillary palps (Ganguly, 2003).

Domains controlling cell polarity and proliferation in the Drosophila tumor suppressor Scribble

Cell polarity and cell proliferation can be coupled in animal tissues, but how they are coupled is not understood. In Drosophila imaginal discs, loss of the neoplastic tumor suppressor gene scribble, which encodes a multidomain scaffolding protein, disrupts epithelial organization and also causes unchecked proliferation. Using an allelic series of mutations along with rescuing transgenes, domain requirements for polarity, proliferation control, and other Scrib functions have been identified. The leucine-rich repeats (LRR) tether Scrib to the plasma membrane, are both necessary and sufficient to organize a polarized epithelial monolayer, and are required for all proliferation control. The PDZ domains, which recruit the LRR to the junctional complex, are dispensable for overall epithelial organization. PDZ domain absence leads to mild polarity defects accompanied by moderate overproliferation, but the PDZ domains alone are insufficient to provide any Scrib function in mutant discs. A model is suggested in which Scrib, via the activity of the LRR, governs proliferation primarily by regulating apicobasal polarity (Zeitler, 2004).

These results highlight the central role of the LRR in Scrib function. Animals with absent or mutant LRR have phenotypes identical to those entirely lacking Scrib, with dramatic effects on both epithelial polarity and growth control. Of the five evolutionarily conserved protein-protein interaction domains in Scrib, only expression of the LRR can provide polarizing and proliferation-controlling activity, with high levels sufficient to effect nearly full rescue. The LRR is also sufficient to mediate membrane localization, whereas a protein lacking the LRR remains in the cytoplasm. Interestingly, alteration of a conserved leucine in the 10th LRR disrupts all Scrib functions and displaces the protein into the cytoplasm. A related alteration in the 13th LRR of C. elegans (Let-413) also prevents membrane localization and function. However, it is clear that the LRR functions as more than a membrane attachment domain because the Scrib PDZs alone are incapable of rescuing any aspect of the mutant phenotype, even when provided with an exogenous membrane targeting signal (Zeitler, 2004).

How can the LRR, which is broadly localized in the absence of PDZ domains, convey information to specifically polarize the apicobasal axis? It has been suggested that a critical role of Scrib in epithelial polarity is the recruitment of Lgl to the lateral cell cortex. Lgl itself is not highly polarized in its distribution, and while it is displaced from the cortex in scrib null GLC embryos, immunofluorescense reveals that Lgl is indeed cortically localized in LRR-expressing scrib 4 GLC embryos, consistent with proper apicobasal polarization in these animals. Therefore, it appears that membrane-localized LRR can mediate interactions that effect cortical recruitment of Lgl, where Lgl can perform its still unknown activities in regulating protein trafficking (Zeitler, 2004).

A surprising result of these experiments is that epithelia can achieve apicobasal organization in the absence of Scrib PDZ domains. Requirement of the PDZ domains for epithelial polarization was expected because of the frequent occurrence of these motifs in proteins that regulate cell polarity. However, Let-413 PDZ domains are also not required to rescue polarity suggesting that LAP protein PDZ domains may be generally dispensable for organizing the apicobasal axis. This finding has implications for understanding the biological roles of LAP protein PDZ-binding partners. In the case of Scrib, it was found that high level expression of a protein lacking the PDZ domains can provide function similar to low levels of expression of full-length protein. The data thus suggest that under physiological conditions PDZ domain interactions contribute to Scrib function in polarity and proliferation control quantitatively rather than qualitatively (Zeitler, 2004).

The quantitative contribution of the PDZ domains may involve their role in polarizing Scrib along the plasma membrane. Analysis of tagged transgenes suggests that Scrib is localized by a two-part mechanism. In the first step, interactions mediated by the LRR bring the protein to the plasma membrane. In the second step, PDZ domain interactions enrich membrane-bound Scrib at the future site of the SJ. This mechanism mirrors the gradual polarization of Scrib during embryonic development, where Scrib is initially localized homogeneously basolaterally, but becomes focused apicolaterally as the final junctional complex forms. Scrib localization to the SJ thus parallels the maturation of the epithelium. Cell junctions are known to be sites of polarized vesicle trafficking, and proteins that control polarity of the entire epithelial cell membrane are localized to the small perijunctional region. Efficient regulation of cell polarity may require high Scrib levels at this location, with the PDZ domains serving to increase the local concentration of the active LRR (Zeitler, 2004).

The data provide direct evidence of a role for Scrib in SJ formation. Although Scrib and Dlg both localize to the SJ, an unambiguous demonstration that either protein is an SJ component has proven difficult. Physical interactions of SJ components with Scrib or Dlg have not yet been found, and whereas dlg and scrib null mutant discs have no SJs, the dramatic disorganization of these tissues raises the possibility that SJ loss might be secondary to the gross polarity disruption. In this work, scrib mutant discs and embryos were identified that polarize and form normal AJs but nevertheless contain severely disrupted SJs. Interestingly, SJ proteins are zygotically produced and first become concentrated at the apical region of the lateral membrane during embryonic stage 14, when the SJ ultrastructurally appears. By contrast, Scrib and Dlg, which are maternally provided, are enriched in this membrane region at stage 9, long before SJs develop. Scrib may therefore prepattern the site of the future SJ to mediate the subsequent coalescence of other SJ components (Zeitler, 2004).

Loss of scrib from imaginal epithelia causes two major defects: mispolarization, reflected in the ectopic distribution of apical and AJ proteins, and overproliferation, resulting in an enormous increase in cell numbers. A key question is whether these effects, which are seen in all three Drosophila neoplastic tumor suppressor gene mutants, are independent or if they are causally interrelated. The independent model posits the existence of Scrib domains that control proliferation and cell polarity without influencing the other function. By contrast, the interdependent model posits that Scrib acts primarily to regulate apicobasal polarity, and that loss of polarity itself disrupts proliferation control (Zeitler, 2004).

Previous views on control of polarity and proliferation by the neoplastic tumor suppressor genes have favored an independent model. These views are influenced by a study of Dlg functional domains, in which deletion of two PDZ domains caused overproliferation within a maintained epithelial monolayer. This finding led to the proposal that Dlg has separable functions in polarity and growth control, with the latter mediated by PDZ domain interactions. Using analogous methods in this study, it was found that deletion of Scrib PDZ domains similarly causes overproliferation in the absence of gross epithelial disorganization. However, the quantitative analysis reveals that PDZ domain deletion does not entirely disrupt, but only partially compromises, proliferation control. Moreover, this proliferation defect can also result from lower levels of full-length Scrib, and in fact can be rescued by expressing high levels of a single Scrib domain, the LRR. These analyses do not rule out the possibility that the PDZ domains, when covalently linked to the LRR, directly contribute to cell proliferation signaling. Nevertheless, any such contribution is likely minor because high levels of LRR alone restore null mutant discs to nearly WT size (Zeitler, 2004).

A formal demonstration of independent functions requires the identification not only of mutant proteins that rescue polarity without restoring proliferation control but also of those that rescue proliferation control without restoring polarity. Such proteins have not been found, using either random mutagenesis or rescue constructs engineered with a knowledge of conserved domains. Both polarity and proliferation control are simultaneously lost when the LRR is mutated, and expression of domains dispensable for polarity (DeltaLRR, myr-DeltaLRR) does not provide any proliferation control, even to a polarized disc. Although the possibility cannot be excluded that for instance specific LRR mutations could create a protein incapable of polarizing tissue but still allows the proper cessation of proliferation, the failure to identify such proteins encourages the consideration of alternative models (Zeitler, 2004).

Several of these data indicate that proliferation control by the Scrib LRR is intimately linked to its polarity-regulating activity. As with C. elegans Let-413, the LRR is both necessary and sufficient to polarize epithelial cells, including embryonic cells that do not overproliferate in scrib mutants. An LRR-specific missense mutation causes a phenotype equivalent to the protein-null condition. By contrast, in hypomorphic mutant animals, moderate disc polarity defects are accompanied by moderate proliferation defects, and in rescue experiments improvements in epithelial architecture accompany reductions in overproliferation. Because hyperproliferation itself is not sufficient to induce mispolarization, the data are consistent with a model in which the primary role of Scrib is to govern cell polarity, and overproliferation is a consequence of polarity disruption (Zeitler, 2004).

The finding that polarized but nevertheless hyperproliferative scrib 4 cells contain specific mislocalized proteins points to a possible mechanism for how polarity disruption could alter proliferation control. The survey revealed misdistributed SJ components, but the altered shapes of scrib 4 (as well as scrib 5 and j7b3) cells suggest that additional proteins may be aberrantly or inefficiently localized. Polarized proteins include growth factor receptors, which are often clustered near cell junctions, and mislocalization of these receptors can lead to altered activity. Although a growth-regulatory protein mislocalized in scrib 4 discs has not yet been identified, the role of the PDZs in Scrib localization to and assembly of the SJ suggests that such a partner might require junctional localization for efficient signaling. A test of this model, and a mechanistic understanding of Scrib function, awaits the identification of Scrib-binding partners (Zeitler, 2004).

The coupling of proliferation control to cell polarity demonstrated in the fly indicates that polarization loss may contribute to human oncogenesis, and not only in neoplastic tumors. It is generally thought that polarity defects are among the last steps during the development of carcinoma in situ, promoting primarily invasion and metastasis. The data suggest that loss of polarity-regulating proteins might also play a role at earlier steps in tumor development by disorganizing specific growth control pathways. Future work will identify, in both flies and mammals, the effectors of cancerous properties altered in depolarized tumors (Zeitler, 2004).

A genetic screen for dominant modifiers of a cyclin E hypomorphic mutation identifies novel regulators of S-phase entry in Drosophila

Cyclin E together with its kinase partner Cdk2 is a critical regulator of entry into S phase. To identify novel genes that regulate the G1- to S-phase transition within a whole animal use was made of a hypomorphic cyclin E mutation, DmcycEJP, which results in a rough eye phenotype. The X and third chromosome deficiencies were screened, candidate genes were tested, and a genetic screen of 55,000 EMS or X-ray-mutagenized flies was carried out for second or third chromosome mutations that dominantly modify the DmcycEJP rough eye phenotype. Focused was placed on the DmcycEJP suppressors, S(DmcycEJP), to identify novel negative regulators of S-phase entry. There are 18 suppressor gene groups with more than one allele and several genes that are represented by only a single allele. All S(DmcycEJP) tested suppress the DmcycEJP rough eye phenotype by increasing the number of S phases in the postmorphogenetic furrow S-phase band. By testing candidates several modifier genes were identifed from the mutagenic screen as well as from the deficiency screen. DmcycEJP suppressor genes fall into five general classes: (1) chromatin remodeling or transcription factors; (2) signaling pathways, and (3) cytoskeletal, (4) cell adhesion, and (5) cytoarchitectural tumor suppressors. The cytoarchitectural tumor suppressors include scribble, lethal-2-giant-larvae (lgl), and discs-large (dlg), loss of function of which leads to neoplastic tumors and disruption of apical-basal cell polarity. The genetic interactions of scribble with S(DmcycEJP) genes were further explored and it was shown that hypomorphic scribble mutants exhibit genetic interactions with lgl, scab (alpha PS3-integrin -- cell adhesion), phyllopod (signaling), dEB1 (microtubule-binding protein -- cytoskeletal), and moira (chromatin remodeling). These interactions of the cytoarchitectural suppressor gene, scribble, with cell adhesion, signaling, cytoskeletal, and chromatin remodeling genes, suggest that these genes may act in a common pathway to negatively regulate cyclin E or S-phase entry (Brumby, 2004).

This work has led to the identification of many genes that when mutated have the ability to dominantly modify the DmcycEJP adult rough eye phenotype and S-phase defect in third instar larval eye imaginal discs. In addition to genes already known to be regulators of Drosophila cyclin E or G1-S progression [such as E2F1; retinoblastoma (Rbf); ago (cdc4) encoding a protein involved in Cyclin E degradation; the EGF receptor pathway genes Egfr and Ras85D, which act to promote Cyclin E protein accumulation, and Hh signaling pathway genes, which act to promote cyclin E transcription], this screen led to the identification of many novel cyclin E interactors. This study mainly concentrated on the suppressors of DmcycEJP, although from the deficiency screen and specifically testing candidates, axin (an inhibitor of Wg signaling), rho1, and crumbs as enhancers of DmcycEJP, which therefore may act as novel positive regulators of G1-S progression, were identified. The suppressors of DmcycEJP identified include the following classes: (1) chromatin remodeling genes brm, mor, Trl, or the transcription factor Zn72D; (2) signaling pathway genes phyl, sina, trio, Abl, RpS6, wg and Wg pathway effectors dsh and arm; (3) genes encoding cytoskeletal proteins dEB1 (encoding a microtubule-binding protein) and expanded (encoding a FERM domain cytoskeletal protein and hyperplastic tumor suppressor); (4) genes encoding cell adhesion proteins scab (encoding an alpha-integrin), cadN (N-Cadherin), shg (E-Cadherin), and fat (encoding an atypical-cadherin and hyperplastic tumor suppressor); and (5) cytoarchitectural tumor suppressor genes scribble, lgl, and dlg, required for apical-basal cell polarity and cell proliferation inhibition. While some of these genes (brm, mor, expanded, fat, scribble, and lgl) have been previously shown or implicated to play a role in negatively regulating G1-S, a potential role for Trl, Znf72D, phyl, sina, trio, Abl, RpS6, wg, dsh, arm, dEB1, scab, cadN, and shg in inhibiting G1-S progression in Drosophila is novel. Further studies are required to determine whether Abl, RpS6, wg, dsh, arm, and shg do indeed suppress DmcycEJP by acting at the S-phase level and to understand the mechanism by which these genes act in G1-S regulation. The identification of novel classes of presumptive negative regulators of cyclin E or G1-S progression highlights the power of Drosophila whole-animal genetics as a tool for revealing new cell proliferation pathways (Brumby, 2004).

Loss of cell polarity drives tumor growth and invasion through JNK activation in Drosophila

Apparent defects in cell polarity are often seen in human cancer. However, the underlying mechanisms of how cell polarity disruption contributes to tumor progression are unknown. Using a Drosophila genetic model for Ras-induced tumor progression, a molecular link has been shown between loss of cell polarity and tumor malignancy. Mutation of different apicobasal polarity genes activates c-Jun N-terminal kinase (JNK) signaling and downregulates the E-cadherin/β-catenin adhesion complex, both of which are necessary and sufficient to cause oncogenic RasV12-induced benign tumors in the developing eye to exhibit metastatic behavior. Furthermore, activated JNK and Ras signaling cooperate in promoting tumor growth cell autonomously, since JNK signaling switches its proapoptotic role to a progrowth effect in the presence of oncogenic Ras. The finding that such context-dependent alterations promote both tumor growth and metastatic behavior suggests that metastasis-promoting mutations may be selected for based primarily on their growth-promoting capabilities. Similar oncogenic cooperation mediated through these evolutionarily conserved signaling pathways could contribute to human cancer progression (Igaki, 2006).

Most human cancers originate from epithelial tissues. These epithelial tumors, except for those derived from squamous epithelial cells, normally exhibit pronounced apicobasal polarity. However, these tumors commonly show defects in cell polarity as they progress toward malignancy. Although the integrity of cell polarity is essential for normal development, how cell polarity disruption contributes to the signaling mechanisms essential for tumor progression and metastasis is unknown. To address this, a recently established Drosophila model of Ras-induced tumor progression triggered by loss of cell polarity has been used. This fly tumor model exhibits many aspects of metastatic behaviors observed in human malignant cancers, such as basement membrane degradation, loss of E-cadherin expression, migration, invasion, and metastatic spread to other organ sites (Pagliarini, 2003). In the developing eye tissues of these animals, loss of apicobasal polarity is induced by disruption of evolutionarily conserved cell polarity genes such as scribble (scrib), lethal giant larvae (lgl), or discs large (dlg), three polarity genes that function together in a common genetic pathway, as well as other cell polarity genes such as bazooka, stardust, or cdc42. Oncogenic Ras (RasV12), a common alteration in human cancers, causes noninvasive benign overgrowths in these eye tissues (Pagliarini, 2003). Loss of any one of the cell polarity genes somehow strongly cooperates with the effect of RasV12 to promote excess tumor growth and metastatic behavior. However, on their own, clones of scrib mutant cells are eliminated during development in a JNK-dependent manner; expression of RasV12 in these mutant cells prevents this cell death (Igaki, 2006).

To better quantify the metastatic behavior of tumors in different mutant animals, the analysis focused on invasion of the ventral nerve cord (VNC), a process in which tumor cells leave the eye-antennal discs and optic lobes (the areas where they were born) and migrate to and invade a different organ, the VNC. It was further confirmed that the genotypes associated with the invasion of the VNC in this study also resulted in the presence of secondary tumor foci at distant locations, although the number and size of these foci were highly variable (Pagliarini, 2003; Igaki, 2006).

In analyzing the global expression profiles of noninvasive and invasive tumors induced in Drosophila developing eye discs, it was observed that expression of the JNK phosphatase puckered (puc) was strongly upregulated in the invasive tumors. Upregulation of puc represents activation of the JNK pathway in Drosophila. Therefore an enhancer-trap allele, puc-LacZ, was used to monitor the activation of JNK signaling in invasive tumor cells. Strong ectopic JNK activation was present in invasive tumors, while only a slight expression of puc was seen in restricted regions of RasV12-induced noninvasive overgrowth. Intriguingly, more intense JNK activation was seen in tumor cells located in the marginal region of the eye-antennal disc and tumor cells invading the VNC. Analysis of clones of cells with a cell polarity mutation alone revealed that JNK signaling was activated by mutation of cell polarity genes. Notably, JNK signaling was not activated in a strictly cell-autonomous fashion. JNK activation in these cells was further confirmed by anti-phospho-JNK antibody staining that detects activated JNK (Igaki, 2006).

To examine the contribution of JNK activation to metastatic behavior, the JNK pathway was blocked by overexpressing a dominant-negative form of Drosophila JNK (BskDN). As previously reported (Pagliarini, 2003), clones of cells mutant for scrib, lgl, or dlg do not proliferate as well as wild-type clones, while combination of these mutations with RasV12 expression resulted in massive and metastatic tumors. Strikingly, inhibition of JNK activation by BskDN completely blocked the invasion of the VNC, as well as secondary tumor foci formation. Drosophila has two homologs of TRAF proteins (DTRAF1 and DTRAF2), which mediate signals from cell surface receptors to the JNK kinase cascade in mammalian systems. It was found that RNAi-mediated inactivation of DTRAF2, but not DTRAF1, in the tumors strongly suppressed their metastatic behavior. Inactivation of dTAK1, a Drosophila JNK kinase kinase (JNKKK), or Hep, a JNKK, also suppressed metastatic behavior. Drosophila has two known cell surface receptors that act as triggers for the JNK pathway, Wengen (TNF receptor) and PVR (PDGF/VEGF receptor). Intriguingly, it was found that RNAi-mediated inactivation of Wengen partially suppressed tumor invasion. Inactivation of PVR, in contrast, did not show any suppressive effect on metastatic behavior. It was also found that the metastatic behavior of RasV12-expressing tumors that were also mutated for one of three other cell polarity genes, bazooka, stardust, or cdc42, was also blocked by BskDN. These data indicate that loss of cell polarity contributes to metastatic behavior by activating the evolutionarily conserved JNK pathway (Igaki, 2006).

Next, whether JNK activation is sufficient to trigger metastatic behavior in RasV12-induced benign tumors was examined. Two genetic alterations can be used to activate JNK in Drosophila. First, JNK signaling can be activated by overexpression of Eiger, a Drosophila TNF ligand. While mammalian TNF superfamily proteins activate both the JNK and NFκB pathways, Eiger has been shown to specifically activate the JNK pathway through dTAK1 and Hep. Indeed, the eye phenotype caused by Eiger overexpression could be reversed by blocking JNK through Bsk-IR (Bsk-RNAi). Second, overexpression of a constitutively activated form of Hep (HepCA) can also activate JNK signaling. However, the eye phenotype caused by HepCA overexpression was only slightly suppressed by Bsk-IR, suggesting that HepCA overexpression may have additional effects other than JNK activation. Therefore Eiger overexpression was used to activate JNK in RasV12-induced benign tumors, and it was found that the RasV12+Eiger-expressing tumor cells did not result in the invasion of the VNC. This indicates that loss of cell polarity must induce an additional downstream effect(s) essential for metastatic behavior. A strong candidate for the missing event is downregulation of the E-cadherin/catenin adhesion complex, since this complex is frequently downregulated in malignant human cancer cells and is also downregulated by loss of cell polarity genes in Drosophila invasive tumors (Pagliarini, 2003). In addition, it has been recently reported that higher motility of mammalian scrib knockdown cells can be partially rescued by overexpression of E-cadherin-catenin fusion protein, suggesting a role of E-cadherin in preventing polarity-dependent invasion. Furthermore, overexpression of E-cadherin blocks metastatic behavior of RasV12/scrib−/− tumors (Pagliarini, 2003), indicating that loss of E-cadherin is essential for inducing tumor invasion in this model. It was found that loss of the Drosophila E-cadherin homolog shotgun (shg), combined with the expression of both RasV12 and Eiger, induced the invasion of the VNC. Intriguingly, loss of shg in RasV12-expressing clones also showed a weak invasive phenotype at lower penetrance. In agreement with the essential role of JNK in tumor invasion, clones of shg−/− cells weakly upregulated puc expression. It was further found that JNK activation in dlg−/− clones is not blocked by overexpression of E-cadherin, suggesting that mechanism(s) other than loss of E-cadherin also exist for inducing JNK activation downstream of cell polarity disruption. The metastatic behavior of RasV12+Eiger/shg−/− tumors was completely blocked by coexpression of BskDN, indicating a cell-autonomous requirement of JNK activation for this process. Furthermore, it was found that loss of the β-catenin homolog armadillo also induced metastatic behavior in RasV12-induced benign tumors. In contrast, overexpression of HepCA in RasV12/shg−/− cells resulted in neither enhanced tumor growth nor metastatic behavior. Together, these results suggest that, although the RasV12+Eiger/shg−/− does not completely phenocopy the effect of RasV12/scrib−/−, activation of JNK signaling and inactivation of the E-cadherin/catenin complex are the downstream components of cell polarity disruption that trigger metastatic behavior in RasV12-induced benign tumors (Igaki, 2006).

Aside from its evolutionarily conserved role in cell migration and invasion, JNK signaling is also a potent activator of cell death in Drosophila and mammals. Although RasV12-expressing tissues showed a weak and restricted activation of JNK at later stages of development, mutation of cell polarity genes in combination with RasV12 expression constitutively activated JNK signaling. Striking acceleration of tumor growth occurred during days 5 and 6, and these tumors outcompeted surrounding wild-type tissues, resulting in a loss of the unlabeled wild-type cells and a dramatic increase in the GFP-expressing mutant tissue. The activated JNK was correlated with this accelerated tumor growth, suggesting that JNK signaling may play a role in tumor growth. Indeed, in addition to blocking metastatic behavior, inactivation of JNK pathway components strongly suppressed the accelerated tumor growth caused by cell polarity disruption. These results reveal that JNK signaling activated by loss of cell polarity also stimulates tumor growth (Igaki, 2006).

Since JNK signaling is required for both tumor growth and invasion, it was next asked whether these two phenotypes are separable processes. To address this, different types of tumors caused by alterations in genes involved in cell proliferation, growth, and cell polarity were analyzed. Day 6 RasV12/scrib−/− tumors showed moderate tumor growth and VNC invasion phenotypes. Loss of the Akt gene, a component of insulin growth signaling, considerably reduced the tumor load of RasV12/scrib−/− animals but did not impair metastatic behavior. In contrast, overexpression of Akt, combined with mutations in both the scrib gene and the lats gene, a potent tumor suppressor, did not cause metastatic behavior despite accelerated tumor growth comparable to RasV12/scrib−/−. In addition, although RasV12/Tsc1−/− mutant cells resulted in extremely large tumors, these tumor cells never exhibited metastatic behavior. These data indicate that tumor growth and invasion are separable processes in this model system (Igaki, 2006).

It was found that JNK signaling is indeed activated in polarity-deficient cells, and acridine orange staining revealed that most of these cells die. Interestingly, ectopic cell death was mostly blocked within clones of polarity-deficient cells also expressing RasV12, despite strong JNK activation. In addition, coexpression of RasV12 and Eiger, a potent inducer of cell death, resulted in accelerated tumor growth, although neither RasV12 alone nor Eiger alone caused dramatic overgrowth. This massive overgrowth was completely blocked by coexpression of BskDN. Moreover, stimulation of JNK signaling by expressing Eiger dramatically enhanced tumor growth of RasV12/shg−/− tissues, although Eiger/shg−/− clones were very small, probably because of cell death of these mutant clones. The accelerated growth of the RasV12+Eiger/shg−/− tumors was again completely blocked by BskDN. Together, these data indicate that, in the context of oncogenic Ras, JNK activation is the primary mediator of tumor growth downstream of cell polarity disruption. These observations suggest that JNK signaling switches its proapoptotic role to a progrowth effect in the presence of oncogenic Ras, and that the dramatic tumor growth is caused by cooperation between oncogenic Ras and JNK signaling (Igaki, 2006).

This study provides a molecular link between loss of cell polarity and tumor malignancy, namely activation of JNK signaling and inactivation of the E-cadherin/catenin complex in the context of oncogenic Ras activation. Although RasV12 alone only induces noninvasive overgrowth, and loss of cell polarity alone results in JNK-mediated cell death, the combination of these two alterations promotes both tumor growth and invasion through oncogenic cooperation. Thus, the tumor-promoting alterations caused by loss of cell polarity do not function alone and rather act as oncogenic Ras modifiers or “oncomodifiers” (Igaki, 2006).

The JNK signaling is essential for a variety of biological processes such as morphogenesis, cell proliferation, migration, invasion, and cell death. Genetic studies in Drosophila have demonstrated that JNK signaling is essential for epithelial cell movements and invasive behavior during normal development. A genetic study in mice revealed that TNF-triggered JNK signaling stimulates epidermal proliferation. These studies suggest that JNK may play an important role in tumorigenesis, tumor growth, and metastasis. Indeed, a substantial body of evidence indicates that JNK activation and c-Jun phosphorylation play important roles in cancer development. In mammalian cell culture systems, Ras acts cooperatively with JNK or c-Jun to enhance cellular transformation. Furthermore, knockin mice expressing a mutant form of c-Jun (JunS63A,S73A) suppress development of skin tumors in response to Ras activation and also block development of intestinal epithelial cancers caused by APC mutation. Moreover, liver-specific inactivation of c-Jun impairs development of chemically induced hepatocellular carcinomas. Furthermore, JNK signaling is activated in many tumor types. On the contrary, however, it has been also shown that JNK functions as a negative regulator for tumor development in Ras/p53-transformed fibroblasts. Thus, the role of JNK signaling seems to be highly dependent on cellular context, and, this study provides the first evidence for a cell-autonomous oncogenic cooperation between JNK and Ras signaling that promotes tumor growth and malignancy (Igaki, 2006).

How is JNK signaling activated? Loss of cell polarity may directly influence activity of a JNK pathway component. Alternatively, cell polarity defects may activate a cell surface receptor that triggers JNK signaling. The genetic analysis of multiple JNK pathway components suggests that the pathway is activated through a cell surface receptor, Wengen. It would be interesting to further investigate whether mislocalization or disregulation of Wengen, which should be normally tightly regulated in polarized epithelial cells, results in stimulation of JNK pathway signaling (Igaki, 2006).

The discovery that metastasis-promoting alterations (i.e., JNK activation) also increase tumor growth may explain why tumor cells acquire such mutations; that is, they primarily provide a selective advantage in tumor growth. Given that cell polarity defects are frequently associated with human tumor malignancy, and that the pathways identified in Drosophila are evolutionarily conserved, similar molecular mechanisms could be involved in human tumor progression. It would be particularly interesting to study these processes in human tumors with high frequencies of Ras mutations. If such processes prove conserved, components of these pathways, especially JNK signaling, could serve as potential therapeutic targets against such cancers (Igaki, 2006).

Basolateral junctions utilize warts signaling to control epithelial-mesenchymal transition and proliferation crucial for migration and invasion of Drosophila ovarian epithelial cells

Fasciclin2 (Fas2) and Discs large (Dlg) localize to the basolateral junction (BLJ) of Drosophila follicle epithelial cells and inhibit their proliferation and invasion. To identify a BLJ signaling pathway a genome-wide screen was performed for mutants that enhance dlg tumorigenesis. Two genes were identified that encode known BLJ scaffolding proteins, lethal giant larvae (lgl) and scribble (scrib), and several not previously associated with BLJ function, including warts (wts) and roughened eye (roe/rotund), which encode a serine-threonine kinase and a transcription factor, respectively. Like scrib, wts and roe also enhance Fas2 and lgl tumorigenesis. Further, scrib, wts, and roe block border cell migration, and cause noninvasive tumors that resemble dlg partial loss of function, suggesting that the BLJ utilizes Wts signaling to repress EMT and proliferation, but not motility. Apicolateral junction proteins Fat (Ft), Expanded (Ex), and Merlin (Mer) either are not involved in these processes, or have highly spatio-temporally restricted roles, diminishing their significance as upstream inputs to Wts in follicle cells. This is further indicated in that Wts targets, CyclinE and DIAP1, are elevated in Fas2, dlg, lgl, wts, and roe cells, but not Fat, ex, or mer cells. Thus, the BLJ appears to regulate epithelial polarity and dynamics not only as a localized scaffold, but also by communicating signals to the nucleus. Wts may be regulated by distinct junction inputs depending on developmental context (Zhao, 2008).

The purpose of this work was to gain greater insight into how the BLJ suppresses epithelial tumorigenesis and invasion by identifying and understanding the function of new genes important for BLJ function. To do so, a genomewide screen was completed for enhancers of dlg, which encodes a scaffolding protein that is a crucial organizer of the BLJ and is a potent repressor of follicle epithelial cell tumorigenesis and invasion. Deficiencies that cumulatively span ∼80% of the autosomes, or 64% of the Drosophila genome were systematically screened. A relatively small number of enhancers, ∼1 per 1000 genes screened, were detected indicating that the screen selected for loci specifically required for dlg function. Thus, the novel dlg enhancer genes that were identified, wts, roe, ebi, as well as at least two genes yet to be identified, are likely to be key collaborators with dlg in suppressing epithelial invasion. The specificity of the interactions between dlg and these enhancers is further indicated in that more than one allele of each gene showed an interaction, in several dlg backgrounds, and the strengths of enhancement were similar to deficiencies defining each locus. wts, roe, and ebi also enhanced Fas2 and lgl, indicating that they are not just important for dlg function, but for the function of the BLJ as a whole. In addition, overexpression of all enhancers except ebi suppressed dlg and Fas2 tumorigenesis, further confirming that the identified genes function in a BLJ network (Zhao, 2008).

BLJ pathway components in the nucleus and their putative relationship to Notch: ebi encodes an F-box protein with WD repeats that promotes protein degradation of specific targets. The failure of ebi overexpression to suppress Fas2 or dlg, and the relatively mild ebi phenotypes (midoogenesis small-nucleus and epithelial-organization defects, but no defects in germinal vesicle localization), suggest that ebi may function in only one of the three branches of BLJ signaling or in a parallel pathway to the BLJ. In the eye, ebi is important for promoting differentiation and inhibiting proliferation, which appear to be separable functions. Thus ebi could enhance Fas2 and dlg tumorigenesis by functioning within the proliferation-repressing branch of the BLJ, or the importance of ebi for differentiation suggests that it could function in the EMT branch of the BLJ or both. In contrast, ebi promotes protein degradation in response to Notch (N) and Drosophila EGF receptor (EgfR) signals, suggesting that it may act in a parallel pathway. Both Ebi and its mammalian homolog, TBL1, function in a corepressor complex through association with nuclear hormone transcriptional corepressor SMRTER/SMRT (Zhao, 2008).

Interestingly, although most N appears to be localized on the apical surface of follicle cells, some N is also localized in BLJs. Thus, it is possible that N localized to the BLJ may signal directly to Ebi. Consistent with this possibility, it was found that all of the genes in the BLJ network share some midoogenesis defects with N, including the small nucleus phenotype, epithelial stratification defects, and mislocalization of the germinal vesicle. The epithelial defects are also reminiscent of N-pathway mutants brainiac and egghead, which are required in the germ line for regulating N that is localized on the apical surface of the follicle cells abutting the germ line. Thus one possibility is that N signaling activity is regulated by its localization to apical vs. basolateral junctions in response to several signaling pathways acting during midoogenesis (Zhao, 2008).

The other modest dlg enhancer that was identified, roe, encodes a Krüppel-family zinc-finger protein that appears to be a transcription factor. Roe is also implicated in Notch signaling and thus may function with Ebi in N-dependent processes as proposed above. However, in contrast to ebi, roe loss caused follicle cell tumors, suggesting that roe may function more directly in a BLJ pathway than ebi. Consistent with a direct role for Roe in BLJ signaling, it was found that roe overexpression suppressed Fas2 and dlg tumorigenesis. Further, as for Fas2, dlg, and wts, roe represses CycE and DIAP1 expression (Zhao, 2008).

Warts was of special interested because of the many similarities observed in the quality and strength of wts and scrib phenotypes, suggesting that they are components in a BLJ signaling pathway, rather than a parallel pathway that cross talks with BLJ signaling. wts encodes a serine/threonine kinase that is an ortholog of human tumor suppressors Lats1 and Lats2, both of which have been linked to highly aggressive breast cancers. The prevailing model for Wts signaling in Drosophila is based on signaling in eye and wing tissue. Wts appears to relay signals from apicolateral junction proteins Ft, Ex, and Mer in wing and eye tissues. However, the results from almost every assay, including early tumor formation, border cell migration, BrdU, PH3, CycE, and DIAP1 expression, indicated little functional overlap between Ft, ex, mer, or mer; ex and wts, thus diminishing the importance of apicolateral Ft-Ex-Mer for Wts activation in follicle cells. The exceptions were that during midoogenesis, Mer is required for border cell migration and Ex is required for the endocycle switch, while both are required for maintenance of epithelial integrity and positioning of the germinal vesicle. However, the involvement of Ex and Mer in these processes are fundamentally distinct from how they act in Wts-dependent processes in other tissues. (1) Ft is not involved; (2) no indication was observed of Ex-Mer synergism; (3) ex, mer, and mer; ex phenotypes are relatively mild when compared to wts. It is concluded that the model for Wts activation in which apicolateral junction proteins Ft, Ex, and Mer play the predominant role cannot be universally applicable in all cell types. Rather, the relative importance of Ex and Mer for Wts regulation appears to depend on developmental context (Zhao, 2008).

Consistent with this proposal, strong functional interdependence and phenotypic similarities were found between Fas2, dlg, lgl, scrib, and wts, thus indicating that the BLJ, not the apicolateral junction, plays the predominant role in Wts regulation during oogenesis. Although genetic evidence alone cannot completely rule out that Wts may act in a parallel pathway to the BLJ and impinge on a set of downstream targets that overlap with those targeted by the BLJ, the following observations favor a model in which the BLJ is more directly involved in Wts regulation (it is noted that these are not mutually exclusive alternatives): (1) over 50 tumor suppressor genes have been identified in Drosophila, but lgl, scrib, and wts were the only strong dlg enhancers identified in this genomewide screen; (2) wts showed strong genetic interactions with Fas2, dlg, and lgl, similar to or stronger than scrib, which encodes a known BLJ protein; (3) wts has early tumor phenotypes similar to dlg partial loss of function and to scrib; (4) wts has the same border cell migration phenotype as scrib; (5) wts has similar small nucleus, epithelial stratification, and germinal vesicle defects as Fas2, dlg, lgl, and scrib; (6) like lgl and scrib, wts overexpression suppressed Fas2 and dlg tumorigenesis; (7) Fas2, dlg, and wts have similar proliferation defects, and (8) Fas2, dlg, and wts similarly repress CycE and DIAP1 expression, which is especially crucial, because CycE and DIAP1 are downstream targets of Wts signaling, and ex and mer had no impact on their expression, contrary to results in other tissues. Thus, the data strongly indicate that the BLJ signals through Wts, and may impinge on Roe in the nucleus, thus suggesting the first BLJ signaling pathway in animal cells. This implies that the BLJ not only acts as a localized scaffold, but also signals to the nucleus to control gene expression, both of which cooperate to regulate epithelial polarity and dynamics (Zhao, 2008).

How can these results in follicle cells, which suggest that Wts acts predominantly downstream of the BLJ, be reconciled with findings in eye tissue, which indicate that Wts acts downstream of the apicolateral junction? Interestingly, the genetic data in the eye suggest that Ft, Ex, and Mer cannot account for all of the signals that activate Wts, because wts overgrowth and tissue disorganization phenotypes are more severe than ft or mer; ex. On the basis of these findings in follicle cells, it is possible that Wts activation in the eye requires additional input from the BLJ. This possibility may have been overlooked thus far because dlg does not appear to have an overgrowth phenotype in the eye. dlg may be essential for additional functions in the eye that are epistatic to its tumor suppressor function, thus preventing loss of cells from the epithelium that could mask an overgrowth phenotype. Consistent with this, when activated Rasv12 is combined with dlg loss, dramatic tumors develop that are larger and more invasive than those produced by Rasv12 alone (Zhao, 2008).

In contrast, Dlg may have a diminished role in Wts signaling in the eye, much as the evidence indicates a diminished role for Ex and Mer in Wts signaling in the ovary. According to this model, Wts receives predominant input from distinct lateral junctions depending on tissue context. One distinction is that ovarian follicle cells are derived from a mesodermal lineage, while the eye and wing tissues are from ectodermal lineages. Further, many genes that disrupt apical-basal polarity and epithelial morphology have only subtle phenotypes in the eye by comparison to the ovary or embryo. Finally, the follicular epithelium requires input from junctions on all three follicle cell surfaces, lateral, apical, and basal, whereas most epithelia require only two, lateral and apical or basal. Thus, ovarian and imaginal tissues are likely to organize signaling pathways acting downstream of epithelial junctions in similar, yet fundamentally different ways to meet the unique organizational requirements of their cell-tissue morphologies. Some or all of these differences may contribute to the suggested specificity observed in Wts signaling downstream of BLJs in follicle cells. In general, these findings raise the possibility for future investigation that depending on the cell-tissue morphologies of a given organ, one lateral junction may play a predominant organizational role, and Wts signaling may act as a universal signaling adapter for mediating contact inhibition from that junction (Zhao, 2008).

An especially interesting aspect of Mer and Ex function that was uncovered in follicle cells is that it appears to be restricted to predominantly postmitotic, differentiated cells, in contrast to the role of Mer and Ex in other tissues. Further, given the absence of an involvement of Ft and lack of Mer-Ex synergism it is concluded that if Mer and Ex would be involved in Wts activation in follicle cells, they would have to function via a fundamentally distinct mechanism than in other tissues. It is proposed that during early oogenesis, the BLJ alone may provide the predominant input to Wts. Then, during midoogenesis, Ex and Mer may become involved in novel interactions with Dlg or other components of the BLJ to activate Wts in spatiotemporally distinct populations of differentiating cells to help achieve their unique developmental functions (Zhao, 2008).

How do wts, scrib, and roe promote motility? It is proposed that Scrib, Wts, and Roe are all crucially involved in EMT. In EMT, cells (1) loose apical-basal polarity and become mesenchymal-like, and (2) adopt a polarity conducive to movement. scrib, wts, and roe cells clearly lose epithelial polarity and become mesenchymal-like as indicated by their rounded morphology and lateralized phenotype. However, scrib, wts, and roe tumors do not invade, and scrib, wts, and roe border cells do not move, suggesting that the second aspect of EMT, adoption of a polarity conducive to movement, is defective. Consistent with this, mammalian Scrib is required for migration and epithelial wound healing of cultured human breast epithelial cells, and is also required in vivo for wound healing in mice. Human Scrib directs migration by organizing several polarities crucial for migration, including the orientation of the microtubule and Golgi networks and the localization of Cdc42 and Rac1 to the cell's leading edge. Thus Scrib has a conserved function in directed cell migration by organizing a polarity conducive to movement. In mammalian PC12 cells Scrib is in complex with Rac1. Fly Rac1 is essential for border cell migration and invasion of Fas2 and dlg tumors, suggesting that an essential role of Scrib in Rac1 function may be of crucial importance for movement. The apparent conserved role of BLJ proteins in organizing EMT, and both promoting and repressing movement, reemphasizes the suggestion that BLJ proteins do more than merely maintain apical-basal polarity, but rather repress a cellular transformation from epithelial polarity to a mesenchymal, lateralized signature conducive to movement (Zhao, 2008).

How is the function of scrib, wts, and roe in promoting border cell movement consistent with the requirement of Fas2, dlg, and lgl in repressing border cell movement? Further, how do scrib and wts act as enhancers of dlg tumor invasion even though scrib and wts tumors are noninvasive? For border cell movement, Fas2 and dlg mutations not only accelerate movement, but also delay border cell delamination. The delay in border cell delamination suggests that the BLJ normally promotes motility, but this promoting function can be bypassed when the repression of motility branch of the BLJ pathway is simultaneously lost. Cumulative data indicate that scrib, wts, and roe act predominantly within the EMT and proliferation branches of the BLJ pathway, and not the repression of motility branch. It is suggested that without simultaneous loss of the repression of motility branch of the BLJ pathway, scrib and wts border cells cannot bypass the essential requirement for the second step of EMT, thus border cell motility is blocked (Zhao, 2008).

This interpretation is also consistent with the seemingly paradoxical function of scrib and wts as enhancers of dlg tumor invasion, even though Scrib and Wts promote rather than repress border cell movement. The noninvasive scrib and wts tumor phenotypes indicate that they are crucial for repressing the first step of EMT, loss of epithelial polarity and adoption of a lateralized, mesenchymal-like phenotype. It has been suggested that scrib and wts enhance dlg invasive tumorigenesis by increasing the rate at which dlg mutant follicle cells undergo EMT and further facilitate invasion by depressing proliferation control and increasing the number of follicle cells available for movement. Thus, even though scrib and wts are required to promote movement, it is suggested that in dlg; scrib/+ or dlg; wts/+ tumors this requirement can be bypassed because the branch of the BLJ pathway that represses motility is simultaneously disrupted (Zhao, 2008).

The noninvasive tumor phenotypes of scrib and wts are very similar to the phenotypes of dlg mutants that specifically disrupt Dlg SH3 and GuK domains. Thus Scrib and Wts may act specifically downstream of the Dlg SH3 and GuK domains. Consistent with this, Scrib appears to associate with the Dlg GuK domain in neuronal synapses via the linker protein GuK-holder. Further, whereas Fas2, dlg, and lgl cause faster border cell migration, border cell migration is very similar to wild type in the dlg SH3/GuK-specific mutants, suggesting that Dlg SH3/GuK predominantly represses the first step of EMT and proliferation but not motility. On the basis of this specificity, it is suggest that one reason that lgl may be a stronger dlg enhancer than scrib and wts is that lgl represses motility in addition to EMT and proliferation. For example, the de novo tumor formation observed when one copy of lgl, scrib, or wts is removed in dlghf/dlgsw ovaries suggests that a threshold level of BLJ activity essential for maintenance of polarity has been lost. However, the lgl interaction may be much stronger than scrib and wts because lgl additionally represses motility (Zhao, 2008).

Increased expression of CycE and DIAP1, known Wts targets, was observed in Fas2, dlg, lgl, scrib, wts, and roe cells. Thus the importance of CycE for proliferation control, and DIAP1 for control of EMT and motility, suggests that part of the mechanism by which Fas2-Dlg represses tumorigenesis is through activating Wts signaling. DIAP1 is in a complex with Rac1 and Profilin and enables border cell motility apparently by promoting actin turnover. Further, in the embryo, DIAP1 loss leads to Dlg cleavage and cellular rounding and dispersal. Too much DIAP1 also appears to be deleterious to movement, because targeted overexpression of DIAP1 specifically in border cells slows their migration (data not shown). Thus maintaining the proper balance of DIAP1 is critical for directed movement, and it may be part of the mechanism by which Scrib and Wts influence border cell movement, suggesting that interaction with Dlg and Rac1 may be another level at which Scrib regulates EMT and movement, consistent with the possibility that it functions downstream of Scrib and Wts in follicle cells to repress both EMT and proliferation (Zhao, 2008).

In contrast to the strong enhancement of dlg by scrib, Fas2 was only weakly enhanced by scrib. Given the complexity of coordinating EMT, proliferation, and motility within an epithelial field, perhaps the simplest model is that multiple Dlg complexes reside within the BLJ, each with a distinct set of ligands that control one or more morphogenetic activities (Zhao, 2008).

Another interesting difference in the enhancement of dlg and Fas2 by lgl, scrib, wts, and roe was that they all enhanced both dlg tumorigenesis and invasion, but only enhanced Fas2 tumorigenesis, without invasion. An important difference between these experiments may be that in Fas2null follicle cells, Dlg is missing Fas2 as a ligand, whereas in dlghf/dlgsw, dlghf/dlgip20, and dlghf/dlglv55 follicle cells, Fas2 is localized at sites of contact between follicle cells in both the native epithelium and in streams of invading cells, suggesting that Fas2 continues to act as a Dlg ligand in these cells. This is probably an important difference because Fas2-Dlg binding is expected to control the conformation of Dlg. Dlg conformations in turn may specify Dlg intra- and intermolecular interactions that determine the relative balance of EMT, proliferation, and invasion factors that associate with the BLJ scaffold. For example, in neuronal cells intramolecular interactions between Dlg SH3 and GuK domains regulate the strength of intermolecular binding of GuK-holder, which binds Scrib. The SH3-GuK intramolecular interaction is further modulated by intramolecular interactions with PDZ3, which are regulated by intermolecular interactions with neurolignin, a transmembrane ligand for PDZ3 (Zhao, 2008).

On the basis of this molecular model, it is proposed that in the absence of Fas2, Dlg has a distinct conformation that tilts the balance toward EMT and proliferation over invasion, when Lgl, Scrib, Wts, or Roe are reduced. This study has shown that lgl, scrib, wts, and roe are expected to act predominantly downstream of Dlg SH3 and GuK domains to repress EMT and proliferation. Thus, removal of one copy of lgl, scrib, wts, or roe in Fas2 cells may tip the ratio of factors controlling EMT, motility, and proliferation toward derepression of EMT and proliferation, masking the Fas2 requirement for invasion. One possibility is that lgl, scrib, wts, or roe are especially important for expression of a protein in the apicolateral junction, such as Par-3/Bazooka, which is essential for dlg invasion. Consistent with this, Ex upregulation is seen in both dlg and wts clones. Further, lgl enhancement at the lglts permissive temperature showed essentially the opposite trend from Fas2. Rather than enhance tumorigenesis over invasion, removal of one copy of Fas2, dlg, scrib, wts, or roe in lgl egg chambers favored invasion. Thus, it is suggested that tumor invasiveness associated with particular combinations of mutated BLJ proteins may be masked or unmasked on the basis of the balance of activities that are disrupted, rather than disruption of particular activities per se (Zhao, 2008).

In summary, this study has identified the first signaling pathway that acts downstream of the BLJ that specifically controls EMT and proliferation, and important clues have been gained as to how this signaling may be organized. Like the Drosophila follicular epithelium, the human ovarian surface epithelium, which is thought to be the site of origin of most ovarian cancers, is derived from a mesodermal lineage. The data suggest that the BLJ plays an especially crucial role in the follicle cells compared to ectodermal lineages in repressing epithelial invasion and that the follicular epithelium appears to organize signaling from epithelial junctions in distinct ways compared to other epithelia. Given the conservation in the lineage of the fly and human epithelia, and the sensitivity of this screen for detecting molecules important for invasive carcinogenesis, it is proposed that the fly egg chamber may serve as a prototype for identifying early molecular events that are crucial for invasion of human ovarian cancer and possibly other malignancies that remain undetected before they start to invade (Zhao, 2008).

Intrinsic tumor suppression and epithelial maintenance by endocytic activation of Eiger/TNF signaling in Drosophila

Oncogenic alterations in epithelial tissues often trigger apoptosis, suggesting an evolutionary mechanism by which organisms eliminate aberrant cells from epithelia. In Drosophila imaginal epithelia, clones of cells mutant for tumor suppressors, such as scrib or dlg, lose their polarity and are eliminated by cell death. This study shows that Eiger, the Drosophila tumor necrosis factor (TNF), behaves like a tumor suppressor that eliminates oncogenic cells from epithelia through a local endocytic JNK-activation mechanism. In the absence of Eiger, these polarity-deficient clones are no longer eliminated; instead, they grow aggressively into tumors. In scrib clones endocytosis is elevated, which translocates Eiger to endocytic vesicles and leads to activation of apoptotic JNK signaling. Furthermore, blocking endocytosis prevents both JNK activation and cell elimination. These data indicate that TNF signaling and the endocytic machinery could be components of an evolutionarily conserved fail-safe mechanism by which animals protect against neoplastic development (Igaki, 2009).

Clones of cells mutant for Drosophila tumor suppressor genes, such as scrib or dlg, are eliminated from imaginal discs, suggesting an evolutionarily conserved fail-safe mechanism that eliminates oncogenic cells from epithelia. This study reports that this elimination of mutant cells is accomplished by endocytic activation of Eiger/TNF signaling. Eiger is a conserved member of the TNF superfamily in Drosophila, but its physiological function has been elusive. Although ectopic overexpression of Eiger can trigger apoptosis, flies deficient for eiger develop normally and exhibit no morphological or cell death defect. This study shows that Eiger is required for the elimination of oncogenic mutant cells from imaginal epithelia. This not only provides an explanation for previous unexplained observations, but also argues that Eiger is a putative intrinsic tumor suppressor in a fashion similar to mammalian p53 or ATM, which causes no phenotype when mutated, but protects animals as tumor suppressors when their somatic cells are damaged (Igaki, 2009).

The intrinsic tumor suppression found in scrib mutant clones was also observed in dlg mutant clones, suggesting that this is a mechanism triggered by loss of epithelial basolateral determinants. Intriguingly, it was found that mutant clones of salvador, the hippo pathway tumor suppressor, are not susceptible to similar effect of Eiger. These data suggest that the Eiger-JNK pathway behaves as an intrinsic tumor suppressor that eliminates cells with disrupted cell polarity (Igaki, 2009).

It is intriguing that Eiger's tumor suppressor-like function is dependent on endocytosis. The data show that Eiger is translocated to endosomes through endocytosis and activates JNK signaling in these vesicles. Moreover, blocking endocytosis abolishes both JNK activation and Eiger-dependent cell elimination. Endocytic activation of signal transduction has been observed for EGF and β2-adrenergic receptor signaling in mammalian cells. After endocytosis, these ligand/receptor complexes localize to endosomes, where they meet adaptor or scaffold proteins that recruit downstream signaling components. Therefore, the endocytic activation of Eiger/TNF-JNK signaling might also be achieved by the recruitment of its downstream signaling complex to the endosomes, possibly through a scaffold protein that resides in endosomes. Recent studies in Drosophila have shown that components of the endocytic pathway -- vps25, erupted, and avalanche -- function as tumor suppressors (Lu, 2005; Moberg, 2005; Thompson, 2005; Vaccari, 2005). Furthermore, mutations in endocytosis proteins have been reported in human cancers. Thus, deregulation of endocytosis may contribute to tumorigenesis. This study provides new mechanistic insights into the role of endocytosis in tumorigenesis (Igaki, 2009).

Mammalian TNF superfamily consists of at least 19 members. While many have been shown to play important roles in immune responses, hematopoiesis, and morphogenesis, the physiological functions for other members have yet to be determined. Mechanisms that eliminate damaged or oncogenic cells from epithelial tissues are essential for multicellular organisms, especially for long-lived mammals like humans. The tumor suppressor role of Eiger might have evolved for host defense or elimination of dying/damaged cells, such as cancerous cells, very early in animal evolution. Given that components of the Eiger tumor suppressor-like machinery (such as Eiger, endocytic pathway components, and JNK pathway components) are conserved from flies to humans, it is also possible that Eiger and its mammalian counterparts are components of an evolutionarily conserved fail-safe by which animals maintain their epithelial integrity to protect against neoplastic development (Igaki, 2009).

Interaction between RasV12 and scribbled clones induces tumour growth and invasion

Human tumours have a large degree of cellular and genetic heterogeneity. Complex cell interactions in the tumour and its microenvironment are thought to have an important role in tumorigenesis and cancer progression. Furthermore, cooperation between oncogenic genetic lesions is required for tumour development; however, it is not known how cell interactions contribute to oncogenic cooperation. The genetic techniques available in the fruitfly Drosophila melanogaster allow analysis of the behaviour of cells with distinct mutations, making this the ideal model organism with which to study cell interactions and oncogenic cooperation. In Drosophila eye-antennal discs, cooperation between the oncogenic protein RasV12 and loss-of-function mutations in the conserved tumour suppressor scribbled (scrib) gives rise to metastatic tumours that display many characteristics observed in human cancers. This study shows that clones of cells bearing different mutations can cooperate to promote tumour growth and invasion in Drosophila. The RasV12 and scrib- mutations can also cause tumours when they affect different adjacent epithelial cells. This interaction between RasV12 and scrib- clones involves JNK signalling propagation and JNK-induced upregulation of JAK/STAT-activating cytokines, a compensatory growth mechanism for tissue homeostasis. The development of RasV12 tumours can also be triggered by tissue damage, a stress condition that activates JNK signalling. Given the conservation of the pathways examined in this study, similar cooperative mechanisms could have a role in the development of human cancers (Wu, 2010).

Clones of mutant cells marked with green fluorescent protein (GFP) can be generated in the eye-antennal imaginal discs of Drosophila larvae by mitotic recombination. Clones expressing RasV12, an oncogenic form of the Drosophila Ras85D protein, moderately overgrow. Clones mutant for scrib lose apico-basal polarity and die. In contrast, scrib clones simultaneously expressing RasV12 grow into large metastatic tumours. To understand better the cooperation between these two mutations, animals were produced in which cell division after a mitotic recombination event creates two daughter cells: one expressing RasV12 and the other mutant for scrib. Discs containing adjacent RasV12 (GFP-positive) and scrib- clones developed into large tumours, capable of invading the ventral nerve cord. This shows that RasV12 and scrib also cooperate for tumour induction when they occur in different cells. These tumours are referred to as RasV12//scrib- tumours, to denote interclonal oncogenic cooperation and distinguish them from RasV12scrib- tumours, in which cooperation occurs in the same cells intraclonally (Wu, 2010).

This study has used Drosophila to investigate how oncogenic cooperation between different cells can promote tumour growth and invasion. These experiments, addressed to understanding interclonal cooperation in RasV12//scrib- tumours, uncovered a two-tier mechanism by which scrib- cells promote neoplastic development of RasV12 cells: (1) propagation of stress-induced JNK activity from scrib- cells to RasV12 cells; and (2) expression of the JAK/STAT-activating Unpaired cytokines downstream of JNK. These findings, therefore, highlight the importance of cell interactions in oncogenic cooperation and tumour development. It was also shown that stress-induced JNK signalling and epigenetic factors such as tissue damage can contribute to tumour development in flies. Notably, tissue damage caused by conditions such as chronic inflammation has been linked to tumorigenesis in humans. Furthermore, expression of the Unpaired cytokines promotes tumour growth as well as an antitumoural immune response, which parallels the situation in mice and humans. Future research into phenomena such as compensatory growth and interclonal cooperation in Drosophila will provide valuable insights into the biology of cancer (Wu, 2010).

Tumor suppression by cell competition through regulation of the Hippo pathway

Homeostatic mechanisms can eliminate abnormal cells to prevent diseases such as cancer. However, the underlying mechanisms of this surveillance are poorly understood. This study investigated how clones of cells mutant for the neoplastic tumor suppressor gene scribble (scrib) are eliminated from Drosophila imaginal discs. When all cells in imaginal discs are mutant for scrib, they hyperactivate the Hippo pathway effector Yorkie (Yki), which drives growth of the discs into large neoplastic masses. Strikingly, when discs also contain normal cells, the scrib- cells do not overproliferate and eventually undergo apoptosis through JNK-dependent mechanisms. However, induction of apoptosis does not explain how scrib- cells are prevented from overproliferating. This study reports that cell competition between scrib- and wild-type cells prevents hyperproliferation by suppressing Yki activity in scrib- cells. Suppressing Yki activation is critical for scrib- clone elimination by cell competition, and experimental elevation of Yki activity in scrib- cells is sufficient to fuel their neoplastic growth. Thus, cell competition acts as a tumor-suppressing mechanism by regulating the Hippo pathway in scrib- cells (Chen, 2012).

This study shows that tumorigenic scrib- cells are removed from Drosophila imaginal discs by a cell-cell signaling event that suppresses elevated Yki activity in scrib- cells. Previous reports implicated JNK as a mediator of cell competition of scrib- clones, where it induces apoptosis and suppresses proliferation. However, it was not known how JNK prevents scrib- clones from hyperproliferating. This study now provides evidence that JNK prevents scrib- clones from hyperproliferating by regulating the activity of the Hippo pathway effector Yki. First, scrib- clones that do not face cell competition up-regulate Yki activity, which drives their hyperproliferation. Second, when scrib- clones do face cell competition, then JNK signaling prevents the upregulation of Yki activity. Third, experimental up-regulation of Yki activity is sufficient to rescue scrib- clones from being eliminated by cell competition. Fourth, experimental suppression of Yki activity in scrib- clones not subjected to cell competition is sufficient to suppress their hyperproliferation. Therefore, cell competition suppresses up-regulation of Yki activity in scrib- cells, and this suppression is important for the elimination of scrib- clones by cell competition. Previous reports showed that Hippo pathway reporters can be up-regulated in scrib- and lgl− mutant discs and clones and that Yki is required for the overgrowth of scrib-+BskDN cells not subjected to cell competition. However, these studies did not analyze the effects of cell competition on Yki activity in scrib- cells. This analysis now shows that scrib- cells facing cell competition do not up-regulate Yki activity and thereby identifies a mechanism that is critical for the elimination of scrib- cells. Although it was reported that scrib- and lgl clones can upregulate ex-lacZ expression and Yki activity (Chen, 2012).

However, upon quantification it was found that the majority of scrib- clones have normal or reduced levels of ex-lacZ expression, and only a small percentage of scrib- clones have elevated levels of ex-lacZ expression. Clones with elevated ex-lacZ expression were observed mainly in the hinge region of wing discs, which may provide an environment of reduced cell competition. Thus, outcompeted scrib- clones do not have elevated levels of Yki activity. In contrast, when scrib- clones are rescued from cell competition, they show highly elevated levels of ex-lacZ expression. Similarly, discs that are entirely mutant for scrib, thereby creating an environment that does not have competing normal cells, show hyperactivation of Yki. Cell competition thus prevents the hyperactivation of Yki in scrib- clones and turns a potential high-Yki 'supercompeting' scrib- cell into a cell of lower fitness and less resistance to apoptosis. Importantly, scrib- wts- and scrib-+Yki clones show greatly increased growth and survival compared with scrib- clones. These results show that elevated levels of Yki are sufficient to protect scrib- cells from being outcompeted. Thus, if Yki activity already was high in scrib- cells facing cell competition, those cells would not be outcompeted, and overexpression of Yki or loss of wts would not cause such dramatic effects on the survival and growth of scrib- clones. Apparently, Yki levels in scrib- cells facing cell competition are not high enough for these cells to evade cell competition. Thus, the amount of Yki activity in scrib- cells is a critical determinant of whether scrib- clones are eliminated or form tumorous tissue, and the suppression of Yki activity in scrib- clones is important for the elimination of scrib- clones by cell competition (Chen, 2012).

These studies show that JNK activity is required in scrib- cells for the suppression of Yki activity by cell competition. In contrast, JNK signaling can induce Yki activity during regeneration and compensatory proliferation in imaginal discs. Therefore, the effects of JNK signaling on Yki activity in scrib- cells are different from those in normal cells: JNK signaling activates Yki in normal cells promoted to regenerate but suppresses Yki in scrib- cells induced to be eliminated. Interestingly, both these effects are observed in discs with scrib- clones. In scrib- cells, JNK activity suppresses the hyperactivation of Yki, but in neighboring cells that are stimulated to proliferate and compensate for the loss of scrib- cells, the activities of both JNK and Yki are elevated. However, non-cell-autonomous effects on Yki reporters were still observed in egr−/− animals and in discs that ubiquitously inhibited JNK signaling by BskDN. Therefore, JNK-independent signals contribute to the non-cell-autonomous induction of Yki activity around scrib- clones. The regulation of Yki by JNK signaling thus is complex and context dependent and may involve several mechanisms (Chen, 2012).

The observation that wts- scrib- clones overgrow indicates that JNK and Wts function in parallel to regulate Yki or that JNK regulates the Hippo pathway upstream of Wts. JNK can phosphorylate and activate Yap1 to regulate apoptosis in mammalian cells. Notably, the JNK phosphorylation sites of Yap1 are different from the Lats phosphorylation sites, supporting a model in which JNK functions in parallel with Wts to regulate Yki activity. However, it is not known whether the same sites also act to suppress the activity of Yki in other contexts (Chen, 2012).

Although several models have been proposed to explain how cell-cell interactions between scrib- and normal cells lead to the elimination of scrib- clones from epithelia, it was not clear what properties normal cells must possess to perform this tumorsuppressive role. The data demonstrate that for scrib- cells to be eliminated they must be juxtaposed with cells that have higher levels of competitive fitness, not just proper cellular architecture. Overexpression of the Myc or RasV12 oncogenes in scrib- clones increases their fitness. As a result, in scrib- clones cell competition does not suppress Yki activity, which protects these clones from being eliminated. Interestingly, Myc expression also synergizes with loss of scrib to form tumors in mammals, and the data offer a model to explain this phenomenon. In addition to the cell-autonomous hyperproliferation, scrib- cells that are not removed from imaginal discs have profound non-cell-autonomous effects on the Hippo pathway. This non-cell-autonomous Hippo pathway-regulating signal may serve normally as a regenerative growth signal that facilitates the replacement of eliminated or dying cells, such as outcompeted scrib- cells. If scrib- clones are not eliminated efficiently, however, this signal may persist longer than required to restore the tissue, thereby causing overgrowth and deformation of neighboring tissue. Thus, continued residence of tumorigenic cells can stimulate growth beyond that needed for compensation, essentially hijacking the proliferation and regeneration programs of their normal neighbors. Therefore, the non-cell-autonomous activation of Yki by scrib- cells may have important implications for tumor-stromal interactions in human cancers (Chen, 2012).

In summary, it is concluded that cell competition is crucial in suppressing the tumorigenic capacity of scrib- cells and does so by regulating their Yki activity. Loss of this regulation results in overproliferation of both tumorigenic cells and neighboring wild-type cells. Efficient elimination of scrib- clones by cell competition prevents Yki-fueled overgrowth of mutant cells and prevents them from disrupting proliferation control of their normal neighbors. Thus, this study identified a tumor-suppression mechanism that depends on signaling between normal and tumorigenic cells. These data identify evasion of cell competition as a critical step toward malignancy and illustrate a role for wild-type tissue in preventing the formation of cancers (Chen, 2012).

Drosophila Src regulates anisotropic apical surface growth to control epithelial tube size.

Networks of epithelial and endothelial tubes are essential for the function of organs such as the lung, kidney and vascular system. The sizes and shapes of these tubes are highly regulated to match their individual functions. Defects in tube size can cause debilitating diseases such as polycystic kidney disease and ischaemia. It is therefore critical to understand how tube dimensions are regulated. This study identified the tyrosine kinase Src as an instructive regulator of epithelial-tube length in the Drosophila tracheal system. Loss-of-function Src42 mutations shorten tracheal tubes, whereas Src42 overexpression elongates them. Surprisingly, Src42 acts distinctly from known tube-size pathways and regulates both the amount of apical surface growth and, with the conserved formin dDaam, the direction of growth. Quantitative three-dimensional image analysis reveals that Src42- and dDaam-mutant tracheal cells expand more in the circumferential than the axial dimension, resulting in tubes that are shorter in length-but larger in diameter-than wild-type tubes. Thus, Src42 and dDaam control tube dimensions by regulating the direction of anisotropic growth, a mechanism that has not previously been described (Nelson, 2012).

To define the molecular mechanisms by which Src42 acts, loss-of-function mutations were tested in candidate Src interactors for a short-tracheal phenotype. dDaam (also known as DAAM, Dishevelled associated activator of morphogenesis), a conserved Diaphanous-related formin that has been shown to bind vertebrate Src, showed a mild, but significant, short-tracheal phenotype near the end of embryogenesis (stage 16). Shortening was more apparent after hatching, raising the possibility that the weaker embryonic phenotype was due to maternal dDaam (Matusek, 2006; Matusek, 2008); failure of dDaam maternal/zygotic embryos to cellularize (Matusek, 2008) precludes a direct test of this possibility). Similarly to Src42, dDaam acts autonomously in the tracheal system, because tracheal expression of Flag-tagged dDaam fully rescued the shortened dorsal trunk of dDaam mutants. Remarkably, despite only causing modest reductions in dorsal-trunk length at embryonic stage 16, zygotic dDaam mutations were still able to completely suppress the overelongation of septate-junction, aECM, apico-basal and PCP mutants. Further, whereas Src42; scrib mutants had gross embryonic defects, dDaam; scrib mutants underwent largely normal development and had trachea with the dDaam short-tracheal phenotype. Thus, dDaam seems to act downstream of or in parallel to all characterized control pathways for tracheal-tube size (Nelson, 2012).

Differential regulation of the Hippo pathway by adherens junctions and apical-basal cell polarity modules

Adherens junctions (AJs) and cell polarity complexes are key players in the establishment and maintenance of apical-basal cell polarity. Loss of AJs or basolateral polarity components promotes tumor formation and metastasis. Recent studies in vertebrate models show that loss of AJs or loss of the basolateral component Scribble (Scrib) cause deregulation of the Hippo tumor suppressor pathway and hyperactivation of its downstream effectors Yes-associated protein (YAP) and Transcriptional coactivator with PDZ-binding motif (TAZ), homologs of Drosophila Yorkie. However, whether AJs and Scrib act through the same or independent mechanisms to regulate Hippo pathway activity is not known. This study dissects how disruption of AJs or loss of basolateral components affect the activity of the Drosophila YAP homolog Yorkie (Yki) during imaginal disc development. Surprisingly, disruption of AJs and loss of basolateral proteins produced very different effects on Yki activity. Yki activity was cell-autonomously decreased but non-cell-autonomously elevated in tissues where the AJ components E-cadherin (E-cad) or α catenin (α-cat) were knocked down. In contrast, scrib knockdown caused a predominantly cell-autonomous activation of Yki. Moreover, disruption of AJs or basolateral proteins had different effects on cell polarity and tissue size. Simultaneous knockdown of α-cat and scrib induced both cell-autonomous and non-cell-autonomous Yki activity. In mammalian cells, knockdown of E-cad or α-cat caused nuclear accumulation and activation of YAP without overt effects on Scrib localization and vice versa. Therefore, these results indicate the existence of multiple, genetically separable inputs from AJs and cell polarity complexes into Yki/YAP regulation. (Yang, 2014).

This report addresses the effects of AJs and basolateral cell polarity determinants on the activity of the Hippo pathway in Drosophila imaginal discs. Knockdown of AJs and basolateral components both induced ectopic activation of Yki. However, knockdown of AJs and basolateral proteins had strikingly different effects on Yki. Disruption of the basolateral module induced mainly a cell-autonomous increase in Yki activity, whereas knockdown of AJs caused non-autonomous induction of Yki reporters. Therefore, these data identify and genetically uncouple multiple different molecular pathways from AJs and the basolateral module that regulate Yki activity (Yang, 2014).

These studies further show that knockdown of AJs induces cell-autonomous reduction of Yki activity and causes cell death and decreased size of Drosophila imaginal discs. Likewise, E-cad and :alpha;-cat mutant clones do not survive in imaginal discs. This effect may be mediated by LIM domain proteins of the Zyxin and Ajuba subfamilies, which regulate Hippo signaling by directly inhibiting Wts/Lats kinases and by interacting with Salvador (Sav), an adaptor protein that binds to the Hpo/MST kinases. A recent report shows that α-Cat recruits Ajuba and indirectly Wts to AJs and loss of Ajuba leads to activation of Wts and hence phosphorylation and inhibition of Yki and diminished tissue size. Thus, α-cat mutant cells may inactivate Yki because they lose Ajuba function (Yang, 2014).

In contrast, in mammalian systems, several in vivo and in vitro studies have shown the opposite effect on Hippo signaling upon AJ disruption; knockdown of E-cad or α-cat caused an increase in cell proliferation and nuclear accumulation of YAP, and conditional knockout of α-cat in mouse skin cells caused tumor formation and elevated nuclear YAP staining. This suggests that AJ components have a tumor suppressor function in mammals. The observation that Scrib is mislocalized upon disruption of AJs in several different mammalian cell lines suggested that YAP activation could be due to the concomitant disruption of the basolateral module. However, the finding that acute disruption of AJs can cause YAP activation without disrupting Scrib localization and vice versa indicates that AJs and the basolateral module also act independently on the Hippo pathway in mammalian cells. In mammalian cells, α-Cat forms a complex with YAP and 14-3-3 proteins, thereby sequestering phosphorylated YAP at the plasma membrane. However, α-Cat may function as a tumor suppressor only in epidermal stem cells, as conditional deletion of α-cat in differentiated cells only caused a mild phenotype with no overgrowth and tumor formation. Therefore, it is possible that the negative regulation of YAP by α-Cat is cell type-specific, although further testing is required to fully address this issue (Yang, 2014).

The non-cell-autonomous effect of AJ knockdown on the Hippo pathway is an intriguing phenomenon. Several groups reported non-autonomous effects on the Hippo pathway in Drosophila in other mutant conditions. Disrupting the expression gradients of the atypical Cadherin Dachsous or that of its regulator Four-jointed, clones of cells mutant for the tumor suppressor genes vps25 or hyperplastic discs (hyd) , clones of cells overexpressing Src64, or overexpression of the proapoptotic gene reaper or the JNK signaling ligand eiger all cause non-autonomous activation of Yki. This non-autonomous activation of Yki may be part of a regenerative response that stimulates cell proliferation in cells neighboring tissue defects. The signals that activate Yki in these situations are not known, nor is it known whether these mutant conditions activate the same or different signaling mechanisms. The non-autonomous activation of Yki around cells with AJ knockdown may be mediated by changes in mechanical forces. AJs are important for maintaining tension between cells across epithelia, and disruption of AJs leads to an imbalance of apical tension. Mechanical forces are known to regulate the Hippo pathway, and YAP/TAZ act as mediators of mechanical cues from the cellular microenvironment such as matrix stiffness. In particular, the Zyxin and Ajuba family LIM domain proteins can act as sensors of mechanical forces and may be involved in the non-autonomous activation of Yki. The effects on Hippo signaling of solely changing Zyxin and Ajuba may not be as strong as those described here, and these proteins may thus cooperate with other molecular conduits to regulate the activity of the Hippo pathway in response to changes in AJ strength. Unraveling these mechanisms will provide important new insights into understanding how cells interact with neighboring cells to regulate proliferation, apoptosis, and the Hippo pathway (Yang, 2014).

It is currently unknown whether AJs also exert non-autonomous effects on the Hippo pathway in mammalian tissues. Amphiregulin, an EGF ligand, is a downstream target of YAP and can induce non-cell-autonomous cell proliferation through EGFR signaling. However, it is not known whether YAP itself is activated non-cell-autonomously to contribute to the hyper-proliferation phenotypes observed upon disruption of AJs in vivo and in vitro. It will be interesting to determine whether AJs and other cell-cell signaling mechanisms also have non-cell-autonomous effects on the activity of YAP in mammalian tissues, for example during regeneration (Yang, 2014).

Finally, the apical proteins aPKC and Crb modulate the activity of the Hippo pathway, and many Hippo pathway components are apically localized, which is important for their activity. The data presented in this study add to these findings. Disruption of AJs causes reduced Yki activity, despite the fact that Crb and Mer are mislocalized. Thus, AJs and cell polarity components regulate Yki activity through multiple, genetically separable inputs. It will be interesting to decipher all of the different underlying molecular mechanisms of how AJs and basolateral proteins regulate the Hippo pathway and how these mechanisms evolved in Drosophila and in mammals (Yang, 2014).

The Drosophila TNF receptor Grindelwald couples loss of cell polarity and neoplastic growth

Disruption of epithelial polarity is a key event in the acquisition of neoplastic growth. JNK signalling is known to play an important part in driving the malignant progression of many epithelial tumours, although the link between loss of polarity and JNK signalling remains elusive. In a Drosophila genome-wide genetic screen designed to identify molecules implicated in neoplastic growth, this study identified grindelwald (grnd; CG10176), a gene encoding a transmembrane protein with homology to members of the tumour necrosis factor receptor (TNFR) superfamily. This study shows that Grnd mediates the pro-apoptotic functions of Eiger (Egr), the unique Drosophila TNF, and that overexpression of an active form of Grnd lacking the extracellular domain is sufficient to activate JNK signalling in vivo. Grnd also promotes the invasiveness of RasV12/scrib-/- tumours through Egr-dependent Matrix metalloprotease-1 (Mmp1) expression. Grnd localizes to the subapical membrane domain with the cell polarity determinant Crumbs (Crb) and couples Crb-induced loss of polarity with JNK activation and neoplastic growth through physical interaction with Veli (also known as Lin-7). Therefore, Grnd represents the first example of a TNFR that integrates signals from both Egr and apical polarity determinants to induce JNK-dependent cell death or tumour growth (Andersen, 2015).

A genome-wide screen was carried to identify molecules that are required for neoplastic growth. The condition used for this screen was the disc-specific knockdown of avalanche, also known as syntaxin 7), a gene encoding a syntaxin that functions in the early step of endocytosis2. avl-RNAi results in ectopic Wingless (Wg) expression, neoplastic disc overgrowth, and a 2-day delay in larva-to-pupa transition. A collection of 10,100 transgenic RNA interference (RNAi) lines were screened for their ability to rescue the pupariation delay, and 121 candidate genes were identified. Interestingly, only eight candidate genes also rescued ectopic Wg expression and neoplastic overgrowth. These included five lines targeting core components of the JNK pathway (Bendless, Tab2, Tak1, Hemipterous and Basket. Using a puckered enhancer trap (puc-lacZ) as a readout for JNK activity, it was confirmed that JNK signalling is highly upregulated in avl-RNAi discs. One of the remaining lines targets CG10176, a gene encoding a transmembrane protein. Reducing expression of CG10176 by using two different RNAi lines was as efficient as tak1 silencing to restore normal Wg pattern and suppresses JNK signalling and neoplastic growth in the avl-RNAi background. Sequence analysis of GC10176 identified a cysteine-rich domain (CRD) in the extracellular part with homology to vertebrate TNFRs harbouring a glycosphingolipid-binding motif (GBM) characteristic of many TNFRs including Fas. CG10176 was named grindelwald (grnd) , after a village at the foot of Eiger, a Swiss mountain that lent its name to the unique Drosophila TNF, Egr. Immunostaining and subcellular fractionation of disc extracts confirmed that Grnd localizes to the membrane. Moreover, co-immunoprecipitation experiments showed that both Grnd full-length and Grnd-intra, a form lacking its extracellular domain, directly associate with Traf2, the most upstream component of the JNK pathway. This interaction is disrupted by a single amino acid substitution within a conserved Traf6-binding motif (human TRAF6 is the closest homologue to Traf2. Overexpression of Grnd-intra, but not full-length Grnd, is sufficient to induce JNK signalling, ectopic Wg expression and apoptosis, and Grnd-intra-induced apoptosis is efficiently suppressed in a hep (JNKK) mutant background, confirming that Grnd acts upstream of the JNK signalling cascade (Andersen, 2015).

The Drosophila TNF Egr activates JNK signalling and triggers cell death or proliferation, depending on the cellular context. Therefore tests were performed to see whether Grnd is required for the small-eye phenotype generated by Egr-induced apoptosis in the retinal epithelium (via Egr overexpression). Inhibition of JNK signalling by reducing tak1 or traf2 expression, or by overexpressing puckered, blocks Egr-induced apoptosis and rescues the small-eye phenotype. In contrast to a previous report, RNAi silencing of wengen (wgn) , a gene encoding a presumptive receptor for Egr, does not rescue the small-eye phenotype. Furthermore, the small-eye phenotype is not modified in a wgn-null mutant background, confirming that Wgn is not required for Egr-induced apoptosis in the eye. By contrast, reducing grnd levels partially rescues the Egr-induced small-eye phenotype, producing a 'hanging-eye' phenotype that is not further rescued in a wgn-knockout mutant background. A similar phenotype was previously reported as a result of non-autonomous cell death induced by a diffusible form of Egr. This suggests that Grnd prevents Egr from diffusing outside of its expression domain. Co-immunoprecipitation experiments show that both full-length Grnd and Grnd-extra, a truncated form of Grnd lacking the cytoplasmic domain, associate with Egr through its TNF-homology domain. Although Grnd-extra can bind Egr, it cannot activate JNK signalling. Therefore, it was reasoned that Grnd-extra expression might prevent both cell-autonomous and non-autonomous apoptosis by trapping Egr and preventing its diffusion and binding to endogenous Grnd. Indeed, GMR-Gal4-mediated expression of grnd-extra fully rescues the Egr small-eye phenotype. To confirm that the removal of Grnd induces Egr-mediated non-autonomous cell death, wing disc clones were generated expressing egr alone, egr + tak1 RNAi, or egr + grnd RNAi. As expected, reducing tak1 levels in egr-expressing clones prevents their elimination by apoptosis. Similarly, reducing grnd levels prevents autonomous cell death, but also induces non-autonomous apoptosis. This suggests that Egr, like its mammalian counterpart TNF-α, can be processed into a diffusible form in vivo whose interaction with Grnd limits the potential to act at a distance. Flies carrying homozygous (grndMinos/Minos) or transheterozygous (grndMinos/Df) combinations of a transposon inserted in the grnd locus express no detectable levels of Grnd protein and are equally resistant to Egr-induced cell death. In addition, grndMinos/Minos mutant flies are viable and display no obvious phenotype, suggesting that Grnd, like Egr, participates in a stress response to limit organismal damage. Collectively, these data demonstrate that Grnd is a new Drosophila TNF receptor that mediates most, if not all, Egr-induced apoptosis (Andersen, 2015).

TNFs probably represent a danger signal produced in response to tissue damage to rid the organism of premalignant tissue or to facilitate wound healing. Disc clones mutant for the polarity gene scribbled (scrib) induce an Egr-dependent response resulting in the elimination of scrib mutant cells by JNK-mediated apoptosis. To test the requirement for Grnd in this process, scrib-RNAi and scrib-RNAi + grnd-RNAi clones obtained 72 h after heat shock induction were compared. As expected, scrib-RNAi cells undergo apoptosis and detach from the epithelium. By contrast, scrib-RNAi clones with reduced grnd expression survive, indicating that Grnd is required for Egr-dependent elimination of scrib-RNAi cells. Similar results were obtained by generating scrib mutant clones in the eye disc (Andersen, 2015).

In both mammals and flies, TNFs are double-edged swords that also have the capacity to promote tumorigenesis in specific cellular contexts. Indeed, scrib minus eye disc cells expressing an activated form of Ras (RasV12) exhibit a dramatic tumour-like overgrowth and metastatic behaviour, a process that critically relies on Egr. RasV12/scrib-/- metastatic cells show a strong accumulation of Grnd and Mmp1, and invade the ventral nerve cord. Primary tumour cells reach peripheral tissues such as the fat body and the gut, where they form micro-metastases expressing high levels of Grnd. Reducing grnd levels in RasV12/scrib-/- clones is sufficient to restore normal levels of Mmp1 and abolish invasiveness in a way similar to that observed in an egr mutant background. Therefore, Grnd is required for the Egr-induced metastatic behaviour of RasV12/scrib-/- tumorous cells. Similarly, reducing grnd, but not wgn levels, strongly suppresses Mmp1 expression in RasV12/dlg-RNAi cells and limits tumour invasion, indicating that Wgn does not have a major role in the progression of these tumours (Andersen, 2015).

Perturbation of cell polarity is an early hallmark of tumour progression in epithelial cells. In contrast to small patches of polarity-deficient cells, for example, scrib mutant clones, organ compartments or animals fully composed of polarity-deficient cells become refractory to Egr-induced cell death and develop epithelial tumours. The formation of these tumours requires JNK/MAPK signalling, but not Egr, suggesting Egr-independent coupling between loss of polarity and JNK/MAPK-dependent tumour growth. In line with these observations, it was noticed that, in contrast to Grnd, Egr is not required to drive neoplastic growth in avl-RNAi conditions. This suggests that, in addition to its role in promoting Egr-dependent functions, Grnd couples loss of polarity with JNK-dependent growth independently of Egr. Disc immunostainings revealed that Grnd co-localizes with the apical determinant Crb in the marginal zone, apical to the adherens junction protein E-cadherin (E-cad) and the atypical protein kinase C (aPKC). In avl-RNAi discs, Grnd and Crb accumulate in a wider apical domain. Apical accumulation of Crb is proposed to be partly responsible for the neoplastic growth induced by avl knockdown, since overexpression of Crb or a membrane-bound cytoplasmic tail of Crb (Crb-intra) mimics the avl-RNAi phenotype. Therefore whether Grnd might couple the activity of the Crb complex with JNK-mediated neoplastic growth was examined. Indeed, reducing grnd levels, but not wgn, in ectopic crb-intra discs suppresses neoplastic growth as efficiently as inhibiting the activity of the JNK pathway. Notably, Yki activation is not rescued in these conditions, illustrating the ability of Crb-intra to promote growth independently of Grnd by inhibiting Hippo signalling through its FERM-binding motif (FBM). Indeed, neoplastic growth and polarity defects induced by a form of Crb-intra lacking its FBM (CrbΔFBM-intra) are both rescued by Grnd silencing. As expected, the size of ectopic crbΔFBM-intra;grnd-RNAi discs is reduced compared to the size of ectopic crb-intra; grnd-RNAi discs (Andersen, 2015).

Crb, Stardust (Sdt; PALS1 in humans), and Pals1-associated tight junction protein (Patj) make up the core Crb complex, which recruits the adaptor protein Veli (MALS1-3 in humans). In agreement with previous yeast two-hybrid data, this study found that Grnd binds directly and specifically to the PDZ domain of Veli through a membrane-proximal stretch of 28 amino acids in its intracellular domain. Grnd localization is unaffected in crb and veli RNAi mutant clones. However, reducing veli expression rescues the patterning defects and disc morphology of ectopic crb-intra mutant cells, suggesting that Grnd couples Crb activity with JNK signalling through its interaction with Veli. Interestingly, aPKC-dependent activation of JNK signalling also depends on Grnd. aPKC is capable of directly binding and phosphorylating Crb, which is important for Crb function. This suggests that aPKC, either directly or through Crb phosphorylation, activates Grnd-dependent JNK signalling in response to perturbation of apico-basal polarity (Andersen, 2015).

These data are consistent with a model whereby Grnd integrates signals from Egr, the unique fly TNF, and apical polarity determinants to induce JNK-dependent neoplastic growth or apoptosis in a context-dependent manner. Recent work reveals a correlation between mammalian Crb3 expression and tumorigenic potential in mouse kidney epithelial cells. The conserved nature of the Grnd receptor suggests that specific TNFRs might carry out similar functions in vertebrates, in which the link between apical cell polarity and tumour progression remains elusive (Andersen, 2015).

The transcriptional response to tumorigenic polarity loss in Drosophila

Loss of polarity correlates with progression of epithelial cancers, but how plasma membrane misorganization drives oncogenic transcriptional events remains unclear. The polarity regulators of the Drosophila Scribble (Scrib) module are potent tumor suppressors and provide a model for mechanistic investigation. RNA profiling of Scrib mutant tumors revealed multiple signatures of neoplasia, including altered metabolism and dedifferentiation. Prominent among these was upregulation of cytokine-like Unpaired (Upd) ligands, which drive tumor overgrowth. This study identified a polarity-responsive enhancer in upd3, which was activated in a coincident manner by both JNK-dependent Fos and aPKC-mediated Yki transcription. This enhancer, and Scrib mutant overgrowth in general, were also sensitive to activity of the Polycomb Group (PcG), suggesting that PcG attenuation upon polarity loss potentiated select targets for activation by JNK and Yki. These results link epithelial organization to signaling and epigenetic regulators that control tissue repair programs, and provide insight into why epithelial polarity is tumor-suppressive (Bunker, 2015).

Interplay among Drosophila transcription factors Ets21c, Fos and Ftz-F1 drives JNK-mediated tumor malignancy

This study defines TF network that triggers an abnormal gene expression program promoting malignancy of clonal tumors, generated in Drosophila imaginal disc epithelium by gain of oncogenic Ras (RasV12) and loss of the tumor suppressor Scribble (scrib1). Malignant transformation of the rasV12scrib1 tumors requires TFs of distinct families, namely the bZIP protein Fos, the ETS-domain factor Ets21c and the nuclear receptor Ftz-F1, all acting downstream of Jun-N-terminal kinase (JNK). Depleting any of the three TFs improves viability of tumor-bearing larvae, and this positive effect can be enhanced further by their combined removal. Although both Fos and Ftz-F1 synergistically contribute to rasV12scrib1 tumor invasiveness, only Fos is required for JNK-induced differentiation defects and Matrix metalloprotease (MMP1) upregulation. In contrast, the Fos-dimerizing partner Jun is dispensable for JNK to exert its effects in rasV12scrib1 tumors. Interestingly, Ets21c and Ftz-F1 are transcriptionally induced in these tumors in a JNK- and Fos-dependent manner, thereby demonstrating a hierarchy within the tripartite TF network, with Fos acting as the most upstream JNK effector. Of the three TFs, only Ets21c can efficiently substitute for loss of polarity and cooperate with Ras(V12) in inducing malignant clones that, like rasV12scrib1 tumors, invade other tissues and overexpress MMP1 and the Drosophila insulin-like peptide 8 (Dilp8). While rasV12ets21c tumors require JNK for invasiveness, the JNK activity is dispensable for their growth. In conclusion, this study delineates both unique and overlapping functions of distinct TFs that cooperatively promote aberrant expression of target genes, leading to malignant tumor phenotypes. (Kulshammer, 2015).

Genome-wide transcriptome profiling in the Drosophila epithelial tumor model has generated a comprehensive view of gene expression changes induced by defined oncogenic lesions that cause tumors of an increasing degree of malignancy. These data allowed discovery of how a network of collaborating transcription factors confers malignancy to RasV12scrib1 tumors (Kulshammer, 2015).

This study revealed that the response of transformed RasV12scrib1 epithelial cells is more complex in comparison to those with activated RasV12 alone with respect to both the scope and the magnitude of expression of deregulated genes. Aberrant expression of more than half of the genes in RasV12scrib1 tumors requires JNK activity, highlighting the significance of JNK signaling in malignancy. Importantly, the tumor-associated, JNK-dependent transcripts cluster with biological functions and processes that tightly match the phenotypes of previously described tumor stages. Furthermore, the RasV12scrib1 transcriptome showed significant overlap (27% upregulated and 15% downregulated genes) with microarray data derived from mosaic EAD in which tumors were induced by overexpressing the BTB-zinc finger TF Abrupt (Ab) in scrib1 mutant clones as well as with a transcriptome of scrib1 mutant wing discs. It is proposed that 429 misregulated transcripts (e.g. cher, dilp8, ets21c, ftz-f1, mmp1, upd), shared among all the three data sets irrespective of epithelial type (EAD versus wing disc) or cooperating lesion (RasV12 or Ab), represent a 'polarity response transcriptional signature' that characterizes the response of epithelia to tumorigenic polarity loss. Genome-wide profiling and comparative transcriptome analyses thus provide a foundation to identify novel candidates that drive and/or contribute to tumor development and malignancy while unraveling their connection to loss of polarity and JNK signaling (Kulshammer, 2015).

In agreement with a notion of combinatorial control of gene expression by an interplay among multiple TFs, this study identified overrepresentation of cis-acting DNA elements for STAT, GATA, bHLH, ETS, BTB, bZIP factors and NRs in genes deregulated in RasV12scrib1 mosaic EAD, implying that transcriptome anomalies result from a cross-talk among TFs of different families. Many of the aberrantly expressed genes contained binding motifs for AP-1, Ets21c and Ftz-F1, indicating that these three TFs may regulate a common set of targets and thus cooperatively promote tumorigenesis. This is consistent with the occurrence of composite AP-1-NRRE (nuclear receptor response elements), ETS-NRRE and ETS-AP-1 DNA elements in the regulatory regions of numerous human cancer-related genes, such as genes for cytokines, MMPs (e.g., stromelysin, collagenase) and MMP inhibitors (e.g., TIMP) (Kulshammer, 2015).

Interestingly, Drosophila ets21c and ftz-f1 gene loci themselves contain AP-1 motifs and qualify as polarity response transcriptional signature transcripts. Indeed, this study has detected JNK- and Fos-dependent upregulation of ets21c and ftz-f1 mRNAs in RasV12scrib1 tumors. While JNK-mediated control of ftz-f1 transcription has not been reported previously, upregulation of ets21c in the current tumor model is consistent with JNK requirement for infection-induced expression of ets21c mRNA in Drosophila S2 cells and in vivo. Based on these data, it is proposed that Ftz-F1 and Ets21c are JNK-Fos-inducible TFs that together with AP-1 underlie combinatorial transcriptional regulation and orchestrate responses to cooperating oncogenes. Such an interplay between AP-1 and Ets21c is further supported by a recent discovery of physical interactions between Drosophila Ets21c and the AP-1 components Jun and Fos (Rhee, 2014). Whether regulatory interactions among AP-1, Ets21c and Ftz-F1 require their direct physical contact and/or the presence of composite DNA binding motifs of a particular arrangement to control the tumor-specific transcriptional program remains to be determined (Kulshammer, 2015).

Importantly, some of the corresponding DNA elements, namely AP-1 and STAT binding sites, have recently been found to be enriched in regions of chromatin that become increasingly accessible in RasV12scrib1 mosaic EAD relative to control. This demonstrates that comparative transcriptomics and open chromatin profiling using ATAC-seq and FAIRE-seq are suitable complementary approaches for mining the key regulatory TFs responsible for controlling complex in vivo processes, such as tumorigenesis (Kulshammer, 2015).

The prototypical form of AP-1 is a dimer comprising Jun and Fos proteins. In mammals, the Jun proteins occur as homo- or heterodimers, whereas the Fos proteins must interact with Jun in order to bind the AP-1 sites. In contrast to its mammalian orthologs, the Drosophila Fos protein has been shown to form a homodimer capable of binding to and activating transcription from an AP-1 element, at least in vitro (Kulshammer, 2015).

The role of individual AP-1 proteins in neoplastic transformation and their involvement in pathogenesis of human tumors remain somewhat elusive. While c-Jun, c-Fos and FosB efficiently transform mammalian cells in vitro, only c-Fos overexpression causes osteosarcoma formation, whereas c-Jun is required for development of chemically induced skin and liver tumors in mice. In contrast, JunB acts as a context-dependent tumor suppressor. Thus, cellular and genetic context as well as AP-1 dimer composition play essential roles in dictating the final outcome of AP-1 activity in tumors (Kulshammer, 2015).

This study shows that, similar to blocking JNK with its dominant-negative form, Bsk, removal of Fos inhibits ets21c and ftz-f1 upregulation, suppresses invasiveness, improves epithelial organization and differentiation within RasV12scrib1 tumors and allows larvae to pupate. Strikingly, depletion of Jun had no such tumor-suppressing effects. It is therefore concluded that in the malignant RasV12scrib1 tumors, Fos acts independently of Jun, either as a homodimer or in complex with another, yet unknown partner. A Jun-independent role for Fos is further supported by additional genetic evidence. Fos, but not Jun, is involved in patterning of the Drosophila endoderm Ÿand is required for expression of specific targets, e.g., misshapen (msn) and dopa decarboxylase (ddc), during wound healing. Future studies should establish whether the JNK-responsive genes containing AP-1 motifs, identified in this study, are indeed regulated by Fos without its 'canonical' partner (Kulshammer, 2015).

The current data identify Fos as a key mediator of JNK-induced MMP1 expression and differentiation defects in RasV12scrib1 tumors. Only Fos inhibition caused clear suppression of MMP1 levels and restoration of neurogenesis within clonal EAD tissue, thus mimicking effects of JNK inhibition. Improved differentiation and reduced invasiveness are, however, not sufficient for survival of animals to adulthood, because interfering with Fos function in RasV12scrib1 clones always resulted in pupal lethality (Kulshammer, 2015).

The systems approach of this paper, followed by genetic experiments, identified Ets21c and Ftz-F1 as being essential for RasV12scrib1-driven tumorigenesis. It was further shown that mutual cooperation of both of these TFs with Fos is required to unleash the full malignancy of RasV12scrib1 tumors (Kulshammer, 2015).

TFs of the ETS-domain family are key regulators of development and homeostasis in all metazoans, whereas their aberrant activity has been linked with cancer. ets21c encodes the single ortholog of human Friend leukemia insertion1 (FLI1) and ETS-related gene (ERG) that are commonly overexpressed or translocated in various tumor types. While FLI1 is considered pivotal to development of Ewing's sarcoma, ERG has been linked to leukemia and prostate cancer. As for Ftz-F1 orthologs, the human liver receptor homolog-1 (LRH-1) has been associated with colonic, gastric, breast and pancreatic cancer, whereas steroidogenic factor 1 (SF-1) has been implicated in prostate and testicular cancers and in adrenocortical carcinoma. However, the molecular mechanisms underlying oncogenic activities of either the ERG/FLI1 or the SF-1/LRH-1 proteins are not well understood (Kulshammer, 2015).

This study shows that removal of Ftz-F1 markedly suppressed invasiveness of RasV12scrib1 tumors, restoring the ability of tumor-bearing larvae to pupate. Additionally, and in contrast to Fos, Ftz-F1 inhibition also partly reduced tumor growth in the third-instar EAD and allowed emergence of adults with enlarged, rough eyes composed predominantly of non-clonal tissue. The reduced clonal growth coincided with downregulation of the well-established Yki target, expanded, implicating Ftz-F1 as a potential novel growth regulator acting on the Hpo/Yki pathway. It is further speculated that reduced viability of RasV12scrib1ftz-f1RNAi clones and induction of non-autonomous compensatory proliferation by apoptotic cells during the pupal stage could explain the enlargement of the adult eyes. The precise mechanism underlying compromised growth and invasiveness of RasV12scrib1ftz-f1RNAi tumors and improved survival of the host remains to be identified (Kulshammer, 2015).

In contrast, effects of Ets21cLONG knockdown in RasV12scrib1 tumors appeared moderate relative to the clear improvement conferred by either Fos or Ftz-F1 elimination. ets21cLONG RNAi neither reduced tumor mass nor suppressed invasiveness, and pupation was rescued only partly. However, unlike ftz-f1RNAi, ets21cLONG RNAi significantly reduced expression of dilp8 mRNA. Based on abundance of Ets21c binding motifs in the regulatory regions of tumor-associated genes and the normalized expression of >20% of those genes upon removal of Ets21c, it is further suggested that Ets21c acts in RasV12scrib1 tumors to fine-tune the tumor gene-expression signature (Kulshammer, 2015).

Dilp8 is known to be secreted by damaged, wounded or tumor-like tissues to delay the larval-to-pupal transition. This study has corroborated the role of JNK in stimulating dilp8 expression in RasV12scrib1 tumor tissue, and has further implicated Ets21c and Fos as novel regulators of dilp8 downstream of JNK. However, the data also show that elevated dilp8 transcription per se is not sufficient to delay metamorphosis. Unlike the permanent larvae bearing RasV12scrib1 tumors, those with RasV12scrib1ftz-f1RNAi tumors pupated despite the excessive dilp8 mRNA. Likewise, pupation was not blocked by high dilp8 levels in larvae bearing EAD clones overexpressing Abrupt. As Dilp8 secretion appears critical for its function, it is proposed that loss of Ftz-F1 might interfere with Dilp8 translation, post-translational processing or secretion (Kulshammer, 2015).

Consistent with the individual TFs having unique as well as overlapping functions in specifying properties of RasV12scrib1 tumors, knocking down pairwise combinations of the TFs had synergistic effects on tumor suppression compared with removal of single TF. This evidence supports the view that malignancy is driven by a network of cooperating TFs, and elimination of several tumor hallmarks dictated by this network is key to animal survival. An interplay between AP-1, ETS-domain TFs and NRs is vital for development. For example, the ETS-factor Pointed has been shown to cooperate with Jun to promote R7 photoreceptor formation in the Drosophila adult eye. In mosquitoes, synergistic activity of another ETS-factor, E74B, with the ecdysone receptor (EcR/USP) promotes vitellogenesis. It is thus proposed that tumors become malignant by hijacking the developmental mechanism of combinatorial control of gene activity by distinct TFs (Kulshammer, 2015).

Despite the minor impact of ets21cLONG knockdown on suppressing RasV12scrib1 tumors, Ets21cLONG is the only one of the tested TFs that was capable of substituting for loss of scrib in inducing malignant clonal overgrowth when overexpressed with oncogenic RasV12 in EAD. While invasiveness of such RasV12ets21cLONG tumors required JNK activity, JNK signaling appeared dispensable for tumor growth. Importantly, the overgrowth of RasV12ets21cLONG tumors was primarily independent of a prolonged larval stage, because dramatic tumor mass expansion was detected already on day 6 AEL. How cooperativity between Ets21cLONG and RasV12 ensures sufficient JNK activity and the nature of the downstream effectors driving tumor overgrowth remain to be determined. In contrast, co-expression of either Ftz-F1 or Fos with RasV12 resulted in a non-invasive, RasV12-like hyperplastic phenotype (Kulshammer, 2015).

Why does Ets21cLONG exert its oncogenic potential while Fos and Ftz-F1 do not? Simple overexpression of a TF may not be sufficient, because many TFs require activation by a post-translational modification (e.g., phosphorylation), interaction with a partner protein and/or binding of a specific ligand. Full activation of Fos in response to a range of stimuli is achieved through hyperphosphorylation by mitogen-activated protein kinases (MAPKs), including ERK and JNK. Indeed, overexpression of a FosN-Ala mutated form that cannot be phosphorylated by JNK was sufficient to phenocopy fos deficiency, indicating that Fos must be phosphorylated by JNK in order to exert its oncogenic function. Consistent with the current data, overexpression of FosN-Ala partly restored polarity of lgl mutant EAD cells. It is therefore conclude that the tumorigenic effect of Fos requires a certain level of JNK activation, which is lacking in EAD co-expressing Fos with RasV12. Nevertheless, the absence of an unknown Fos-interacting partner cannot be excluded (Kulshammer, 2015).

Interestingly, MAPK-mediated phosphorylation also greatly enhances the ability of SF-1 and ETS proteins to activate transcription. Two potential MAPK sites can be identified in the hinge region of Ftz-F1, although their functional significance is unknown. Whether Ets21c or Ftz-F1 requires phosphorylation and how this would impact their activity in the tumor context remains to be determined. Genetic experiments demonstrate that at least the overgrowth of RasV12ets21cLONG tumors does not require Ets21c phosphorylation by JNK (Kulshammer, 2015).

In addition, previous crystallography studies revealed the presence of phosphoinositides in the ligand binding pocket of LHR-1 and SF-1 and showed their requirement for the NR transcriptional activity. Although developmental functions of Drosophila Ftz-F1 seem to be ligand independent, it is still possible that Ftz-F1 activity in the tumor context is regulated by a specific ligand. An effect of Ftz-F1 SUMOylation cannot be ruled out (Kulshammer, 2015).

In summary, this work demonstrates that malignant transformation mediated by RasV12 and scrib loss depends on MAPK signaling and at least three TFs of different families, Fos, Ftz-F1 and Ets21c. While their coordinated action ensures precise transcriptional control during development, their aberrant transcriptional (Ets21c, Ftz-F1) and/or post-translational (Fos, Ftz-F1, Ets21c) regulation downstream of the cooperating oncogenes contributes to a full transformation state. The data implicate Fos as a primary nuclear effector of ectopic JNK activity downstream of disturbed polarity that controls ets21c and ftz-f1 expression. Through combinatorial interactions on overlapping sets of target genes and acting on unique promoters, Fos, Ftz-F1 and Ets21c dictate aberrant behavior of RasV12scrib1 tumors. Although originally described in Drosophila, detrimental effects of cooperation between loss of Scrib and oncogenic Ras has recently been demonstrated in mammalian tumor models of prostate and lung cancer. This study and further functional characterization of complex TF interactions in the accessible Drosophila model are therefore apt to provide important insight into processes that govern cancer development and progression in mammals (Kulshammer, 2015).


EVOLUTIONARY HOMOLOGS

FGF21 Induced by the ASK1-p38 Pathway Promotes Mechanical Cell Competition by Attracting Cells

Cell competition is a social cellular phenomenon in which unfit cells are selectively eliminated to maintain tissue homeostasis. Recent studies have revealed that mechanical forces induce competitive cell-cell interactions in Drosophila. This mechanical cell competition has also been reported to play an important role in mammalian cells, using Madin-Darby canine kidney (MDCK) cells depleted of a polarity regulator Scribble in a tetracycline-inducible manner (scrib(KD) cells). scrib(KD) cells are hypersensitive to crowding due to the lower homeostatic density than wild-type (WT) cells, and in the context of cell competition, scrib(KD) cells are compacted and eliminated by WT cells. Although p38 and p53 are involved in this process, the molecular mechanism by which WT cells recognize and mechanically eliminate scrib(KD) cells remains unclear. This study reports that scrib(KD) cells secrete fibroblast growth factor 21 (FGF21) to drive cell competition. Knockdown of FGF21 in scrib(KD) cells or loss of FGFR1 in WT cells suppresses cell competition, suggesting that WT cells recognize scrib(KD) cells through FGF21. FGF21-containing culture medium of scrib(KD) cells activates cell motility. Moreover, FGF21 promotes the compression and elimination of scrib(KD)) cells by attracting surrounding WT cells. It was also demonstrated that activation of the apoptosis signal-regulating kinase 1 (ASK1)-p38 pathway in scrib(KD) cells induces FGF21 to drive cell competition. These findings reveal a mechanism whereby WT cells mechanically eliminate scrib(KD) cells and propose a new function for FGF21 in cell-cell communication (Ogawa, 2020).

Collective nomenclature for LAP proteins

The first Scribbled-like protein to be characterized, Densin-180, was isolated from the rbat postsynaptic density (Apperson, 1996). This protein has a unique structure, in that it contains a set of leucine-rich repeats (LRRs) as well as a PSD-95/Dlg/ZO-1 (PDZ) domain; these domains are thought to mediate protein-protein interactions. Recently, further proteins containing both types of domains have been isolated from fly, worm, mouse and human. It seems an opportune time to select a collective name for this family of proteins. The name 'LAP (LRR And PDZ domain) proteins' is endorsed for proteins with this structure. The known LAP proteins contain 16 canonical LRRs located at the amino terminus of the protein, as well as a conserved, LRR-like region immediately carboxy-terminal to the LRRs. The LRRs from LAP proteins are more closely related to each other than to LRRs from other proteins. The known LAP proteins also contain either one or four PDZ domains. It may be useful to distinguish between these subfamilies by using the designations 'LAP1' and 'LAP4', respectively. Among eukaryotes for which complete genomic sequence is available, Saccharomyces cerevisiae (in which both LRR and PDZ domains can be found) contains no LAP proteins. C. elegans contains a single LAP1 protein, Let-413 (Legouis, 2000), whereas Drosophila contains both a LAP1 protein, dLAP (GenBank AAF58179), and a LAP4 protein, Scribbled (two alternative splice variants, GenBank AF190774 and AJ271647). Vertebrates are likely to contain at least three LAP proteins, because the LAP1 proteins Densin-180 (Bilder, 2000a) and Erbin (Borg, 2000) are both found in rat, whereas human hScribbled is a LAP4 protein (Bilder, 2000a and Nagase, 1995 -- two variants have been identified: GenBank AF271734 and AF240677). Further vertebrate LAP proteins may be identified as complete genomic sequence becomes available. Although analyses of these proteins are just beginning, one striking feature is their polarized localization along the cell membrane. This localization, in conjunction with the modular arrangement of the protein-protein interaction domains they contain, is consistent with genetic and molecular analyses, indicating that LAP proteins may play a key role in regulating the subcellular distribution of other proteins. Identification and further studies of LAP proteins will reveal what aspects of function, as well as of structure, are conserved within this family (Bilder, 2000c).

Localization C. elegans of DLG-1 requires the basolateral LET-413 protein, a Scrib homolog

Epithelial cells are polarized, with apical and basal compartments demarcated by tight and adherens junctions. Proper establishment of these subapical junctions is critical for normal development and histogenesis. The gene let-413 has a critical role in assembling adherens junctions in Caenorhabditis elegans. In let-413 mutants, adherens junctions are abnormal and mislocalized to more basolateral positions; epithelial cell polarity is affected and the actin cytoskeleton is disorganized. The LET-413 protein contains one PDZ domain and 16 leucine-rich repeats with high homology to proteins known to interact with small GTPases. Strikingly, LET-413 localizes to the basolateral membrane. It is suggested that LET-413 acts as an adaptor protein involved in polarizing protein trafficking in epithelial cells (Legouis, 2000).

The correct assembly of junction components, such as E-cadherin and beta-catenin, into the zonula adherens is fundamental for the function of epithelia, both in flies and in vertebrates. In C. elegans, however, the cadherin-catenin system is not essential for general adhesion, raising the question as to the genetic basis controlling junction morphogenesis in nematodes. dlg-1, the C. elegans homolog of the Drosophila tumor-suppressor gene discs-large, plays a crucial role in epithelial development. DLG-1 is restricted to adherens junctions of all embryonic epithelia, which contrasts with the localization of the Drosophila and vertebrate homologs in septate and tight junctions, respectively. Proper localization of DLG-1 requires the basolateral LET-413 protein (identified as the Drosophila scrib ortholog), but is independent of the cadherin-catenin system. Embryos in which dlg-1 activity is eliminated by RNA-mediated interference fail to form a continuous belt of junction-associated antigens and arrest development. Loss of dlg-1 activity differentially affects localization of proteins normally enriched apically to the zonula adherens. While the distribution of an atypical protein kinase C (PKC-3) and other cytoplasmic proteins (PAR-3, PAR-6) is not affected in dlg-1 (RNAi) embryos, the transmembrane protein encoded by crb-1, the C. elegans homolog of Drosophila crumbs, is no longer concentrated in this domain. In contrast to Drosophila, however, crb-1 and a second crb-like gene are not essential for epithelial development in C. elegans. Together the data indicate that several aspects of the spatial organization of epithelial cells and its genetic control differ between flies, worms, and vertebrates, while others are conserved. The molecular nature of DLG-1 makes it a likely candidate to participate in the organization of a protein scaffold that controls the assembly of junction components into the zonula adherens (Bossinger, 2001).

Mammalian Scrib homologs

An abundant protein of apparent molecular mass 180 kDa has been purified from the postsynaptic density fraction of rat forebrain. Amino acid sequences of three tryptic peptides generated from the protein were obtained. The sequences were used to design a strategy cloning the cDNA encoding the protein by polymerase chain reaction. The open reading frame of the cDNA encodes a novel protein of predicted molecular mass 167 kDa. The protein has been named densin-180. Antibodies raised against the predicted amino and carboxyl sequences of densin-180 recognize a 180 kDa band on immunoblots that is enriched in the postsynaptic density fraction. Immunocytochemical localization of densin-180 in dissociated hippocampal neuronal cultures shows that the protein is highly concentrated at synapses along dendrites. The message encoding densin-180 is brain specific and is more abundant in forebrain than in cerebellum. The sequence of densin-180 contains 17 leucine-rich repeats, a sialomucin domain, an apparent transmembrane domain, and a PDZ domain. This arrangement of domains is similar to that of several adhesion molecules, in particular GPIbalpha, which mediates binding of platelets to von Willebrand factor. It is proposed that densin-180 participates in specific adhesion between presynaptic and postsynaptic membranes at glutamatergic synapses (Apperson, 1996).

The ERBB receptors play a crucial role in morphogenesis and oncogenesis. A new PDZ protein, ERBIN (ERBB2 interacting protein), has been identified that acts as an adaptor for the receptor ERBB2/HER2 in epithelia. ERBIN contains 16 leucine-rich repeats (LRRs) in its amino terminus and a PDZ (PSD-95/DLG/ZO-1) domain at its carboxy terminus, and belongs to a new PDZ protein family. The PDZ domain directly and specifically interacts with ERBB2/HER2. ERBIN and ERBB2/HER2 colocalize to the lateral membrane of human intestinal epithelial cells. The ERBIN-binding site in ERBB2/HER2 has a critical role in restricting this receptor to the basolateral membrane of epithelial cells, as mutation of the ERBIN-binding site leads to the mislocalization of the receptor in these cells. It is suggested that ERBIN acts in the localization and signaling of ERBB2/HER2 in epithelia (Borg, 2000).

Densin-180, a brain-specific protein highly concentrated at the postsynaptic density (PSD), belongs to the LAP [leucine-rich repeats and PSD-95/Dlg-A/ZO-1 (PDZ) domains] family of proteins, some of which play fundamental roles in the establishment of cell polarity. To identify new Densin-180-interacting proteins, a yeast two-hybrid library was screened using the COOH-terminal fragment of Densin-180 containing the PDZ domain as bait, and MAGUIN-1 was isolated as a Densin-180-binding protein. MAGUIN-1, a mammalian homologue of Drosophila connector enhancer of KSR (CNK), is known to interact with PSD-95 and has a short isoform, MAGUIN-2. The Densin-180 PDZ domain binds to the COOH-terminal PDZ domain-binding motif of MAGUIN-1. Densin-180 co-immunoprecipitates with MAGUIN-1 as well as with PSD-95 from the rat brain. In dissociated hippocampal neurones Densin-180 co-localizes with MAGUINs and PSD-95, mainly at neuritic spines. In transfected cells, Densin-180 forms a ternary complex with MAGUIN-1 and PSD-95, whereas no association was detected between Densin-180 and PSD-95 in the absence of MAGUIN-1. MAGUIN-1 forms a dimer or multimer via the COOH-terminal leucine-rich region which is present in MAGUIN-1 but not in MAGUIN-2. Among the PDZ domains of PSD-95, the first is sufficient for interaction with MAGUIN-1. These results suggest that the potential to dimerize or multimerize allows MAGUIN-1 to bind simultaneously to both Densin-180 and PSD-95, leading to the ternary complex assembly of these proteins at the postsynaptic membrane (Ohtakara, 2002).

scribble, discs large and lethal giant larvae encode proteins that regulate cell polarity and have been identified as neoplastic tumour suppressor genes in Drosophila melanogaster. The Drosophila model system was used to provide the first functional evidence that human Scribble (hScrib) can act as a tumour suppressor. hScrib protein displays highly polarized localization in mammalian epithelial cells and colocalizes with mammalian Dlg, similar to D. melanogaster Scribble (DmScrib) distribution in Drosophila epithelium. Furthermore, hScrib can rescue the polarity and tumorous overgrowth defects of scrib mutant Drosophila. hScrib therefore can act as an effective tumour suppressor in vivo, regulating both apical-basal polarity and cellular proliferation in a manner similar to that of DmScrib in Drosophila. These data demonstrate that hScrib is a functional homologue of DmScrib and therefore predict an important role for hScrib in the suppression of mammalian tumorigenesis (Dow, 2003).

In mammals, an example of planar cell polarity (PCP) is the uniform orientation of the hair cell stereociliary bundles within the cochlea. The PCP pathway of Drosophila refers to a conserved signalling pathway that regulates the coordinated orientation of cells or structures within the plane of an epithelium. A mutation in Vangl2, a mammalian homolog of the Drosophila PCP gene Strabismus/Van Gogh, results in significant disruptions in the polarization of stereociliary bundles in mouse cochlea as a result of defects in the direction of movement and/or anchoring of the kinocilium within each hair cell. Similar, but less severe, defects are observed in animals containing a mutation in the LAP protein family gene Scrb1 (homologous with Drosophila scribbled). Polarization defects in animals heterozygous for Vangl2 and Scrb1 are comparable to Vangl2 homozygotes, demonstrating genetic interactions between these genes in the regulation of PCP in mammals. These results demonstrate a role for the PCP pathway in planar polarization in mammals, and identify Scrb1 as a PCP gene (Montcouquiol, 2003).

Circletail is one of only two mouse mutants that exhibit the most severe form of neural tube defect (NTD), termed craniorachischisis. In this disorder, almost the entire brain and spinal cord is affected, owing to a failure to initiate neural tube closure. Craniorachischisis is a significant cause of lethality in humans, yet the molecular mechanisms involved remain poorly understood. This study reports the identification of the gene mutated in circletail (Crc), using a positional cloning approach. This gene, Scrb1, encodes a member of the LAP protein family related to Drosophila scribble, with 16 leucine rich repeats and four PDZ domains. The Crc mutant contains a single base insertion that creates a frame shift and leads to premature termination of the Scrb1 protein. Scrb1 expression closely mirrors the phenotypic defects observed in Crc/Crc mutants. In addition, circletail genetically interacts with the loop-tail mutant, and overlapping expression is demonstrated of Scrb1 with Vangl2, the gene mutated in loop-tail. The identification of the Crc gene further defines the nature of the genetic pathway required for the initiation of neural tube closure and provides an important new candidate that may be implicated in the aetiology of human NTDs (Murdoch, 2003).

The asymmetric distribution of proteins to basolateral and apical membranes is an important feature of epithelial cell polarity. To investigate how basolateral LAP [LRR (for leucine-rich repeats) and PDZ (for PSD-95/Discs-large/ZO-1)] proteins, which play key roles in cell polarity, reach their target membrane, a structure-function study of three LAP proteins was carried out: Caenorhabditis elegans LET-413, human Erbin and human Scribble (hScrib). Deletion and point mutation analyses establish that their LRR domain is crucial for basolateral membrane targeting. This property is specific to the LRR domain of LAP proteins, since the non-LAP protein SUR-8 does not localize at the basolateral membrane of epithelial cells, despite having a closely related LRR domain. Importantly, functional studies of LET-413 in C. elegans show that although its PDZ domain is dispensable during embryogenesis, its LRR domain is essential. These data establish a novel paradigm for protein localization by showing that a subset of LRR domains direct subcellular localization in polarized cells (Legouis, 2003).

Among the cellular properties that are essential for the organization of tissues during animal development, the importance of cell polarity in the plane of epithelial sheets has become increasingly clear in the past decades. Planar cell polarity (PCP) signaling in vertebrates has indispensable roles in many aspects of their development, in particular, controlling alignment of various types of epithelial cells. Disrupted PCP has been linked to developmental defects in animals and to human pathology. Neural tube closure defects (NTD) and disorganization of the mechanosensory cells of the organ of Corti are commonly known consequences of disturbed PCP signaling in mammals. A typical PCP phenotype exists in a mouse mutant for the Sec24b gene, including the severe NTD craniorachischisis, abnormal arrangement of outflow tract vessels and disturbed development of the cochlea. In addition, genetic interaction was observed between Sec24b and the known PCP gene, scribble. Sec24b is a component of the COPII coat protein complex that is part of the endoplasmic reticulum (ER)-derived transport vesicles. Sec24 isoforms are thought to be directly involved in cargo selection, and evidence is presented that Sec24b deficiency specifically affects transport of the PCP core protein Vangl2, based on experiments in embryos and in cultured primary cells (Wansleeben, 2010).

Mammalian Scribble forms a tight complex with the ßPIX exchange factor

Drosophila Scribble is implicated in the development of normal synapse structure and epithelial tissues, but it remains unclear how it plays a role and which process it controls. The mammalian homolog of Scribble, hScrib, has a primary structure and subcellular localization similar to that of its fly homolog, but its function remains unknown. Tandem mass spectrometry was used to identify major components of the hScrib network. It includes ßPIX (also called Cool-1), a guanine nucleotide exchange factor (GEF), and its partner GIT1 (also called p95-APP1), a GTPase activating protein (GAP). ßPIX directly binds to the hScrib PDZ domains, and the hScrib/ßPIX complex is efficiently recovered in epithelial and neuronal cells and tissues. In cerebellar granule cell cultures, hScrib and ßPIX are both partially localized at neuronal presynaptic compartments. Furthermore, hScrib is required to anchor ßPIX at the cell cortex and dominant-negative ßPIX or hScrib proteins can each inhibit Ca2+-dependent exocytosis in neuroendocrine PC12 cells, demonstrating a functional relationship between these proteins. These data reveal the existence of a tight hScrib/ßPIX interaction and suggest that this complex potentially plays a role in neuronal transmission (Audebert, 2004).

Although genetic studies have brought about significant insights into the function of Scribble in Drosophila, it remains unclear how Scribble controls epithelial and neuronal morphogenesis at the molecular level. This study has identified the hScrib-ßPIX-GIT1 complex in a variety of tissues, including brain and intestine, and demonstrated that expression of ßPIX promotes regulated exocytosis and that both GEF activity and membrane localization of ßPIX mediated by hScrib are needed in this process. How might the hScrib-ßPIX-GIT1 complex regulate exocytosis? Like hScrib, ßPIX and GIT1 are multidomain proteins engaged in multiple protein-protein interactions. For example, ßPIX is a known partner for Rac1 and Cdc42, two small GTPases that belong to the Rho protein family and are involved in dynamic reorganization of the cytoskeleton. Furthermore, ßPIX has a GEF activity toward these proteins. Interestingly, ßPIX displaying impaired GEF activity did not exhibit exocytotic activity in PC12 cells, suggesting a role of Rac1 or Cdc42 in this process. Indeed, Rac1 has recently been implicated in the control of fusion competence, and Cdc42 has been implicated in promoting the actin structure at the plasma membrane, two steps in exocytosis. Using GST-hScrib PDZ domains, it was possible to pull down a substantial fraction of Rac1 from high KCl-stimulated PC12 cell extracts. This interaction is specific because Cdc42 was not present in the ßPIX-GIT1 complex bound to the hScrib PDZ domains. In light of these data, it is believed that hScrib acts as a membrane anchor for ßPIX, which can then recruit Rac1 to form a functional complex potentially regulating exocytosis (Audebert, 2004).

Major components of the signaling network associated with hScrib have been characterized and a major role has been demonstrated of the PDZ domains in assembling the hScrib-ßPIX-GIT1 complex. Interestingly, the importance of the hScrib PDZ domains was recently highlighted by the characterization of circletail mice harboring an hScrib gene point mutation that leads to a premature termination of the protein after the second PDZ domain. These mice exhibit a profound neural-tube defect (craniorachischisis) and die soon after birth. Removal of two out of four PDZ domains in hScrib may potentially decrease interaction with the ßPIX-GIT1 complex or affect the association of hScrib with additional effectors such as Vangl2, a protein required for planar-cell polarity (Audebert, 2004).

PIX proteins have been conserved throughout evolution, and recent data have shed light on the role of the PIX homolog (dPIX or rtGEF) in neurons in Drosophila (Parnas, 2001). dpix mutants display abnormal presynaptic vesicle accumulation and morphology in neuromuscular junctions (NMJ), and these findings evoke a potential role for this exchange factor in the regulation of exocytosis in the presynaptic compartment. Interestingly, a similar hypothesis was proposed upon observation of NMJ defects in scribble mutants (Roche, 2002). Together, these data suggest that dpix and scribble might participate in a common pathway devoted to vesicle trafficking at presynaptic sites in flies. Future genetic and biochemical experiments will have to evaluate a potential functional interaction among Scribble, PIX, and GIT1 at the NMJ of flies. Drosophila Scribble plays a key function during epithelial polarization. Conservation of this LAP protein throughout evolution suggests a similar function in vertebrates. The presence of an hScrib-ßPIX complex at epithelial cell-cell junctions implies that it may also function during epithelial polarization (Audebert, 2004).

Scrib interacts with the zyxin-related protein LPP, which shuttles between cell adhesion sites and the nucleus

At sites of cell adhesion, proteins exist that not only perform structural tasks but also have a signaling function. The Lipoma Preferred Partner (LPP) protein is localized at sites of cell adhesion such as focal adhesions and cell-cell contacts, and shuttles to the nucleus where it has transcriptional activation capacity. LPP is a member of the zyxin family of proteins, which contains five members: ajuba, LIMD1, LPP, TRIP6 and zyxin. LPP has three LIM domains (zinc-finger protein interaction domains) at its carboxy-terminus, which are preceded by a proline-rich pre-LIM region containing a number of protein interaction domains. To catch the role of LPP at sites of cell adhesion, an effort was made to identify binding partners of LPP. The tumor suppressor protein Scrib, which is a component of cell-cell contacts, has been identified as interaction partner of LPP. Human Scrib, which is a functional homologue of Drosophila scribble, is a member of the leucine-rich repeat and PDZ (LAP) family of proteins that is involved in the regulation of cell adhesion, cell shape and polarity. In addition, Scrib displays tumor suppressor activity. The binding between Scrib and LPP is mediated by the PDZ domains of Scrib and the carboxy-terminus of LPP. Both proteins localize in cell-cell contacts. Whereas LPP is also localized in focal adhesions and in the nucleus, Scrib could not be detected at these locations in MDCKII and CV-1 cells. Furthermore, Scrib is dispensable for targeting LPP to focal adhesions and to cell-cell contacts, and LPP is not necessary for localizing Scrib in cell-cell contacts. All four PDZ domains of Scrib are dispensable for localizing this protein in cell-cell contacts. In conclusion, an interaction has been described between one of zyxin's family members, LPP, and the tumor suppressor protein Scrib. Both proteins localize in cell-cell contacts. This interaction links Scrib to a communication pathway between cell-cell contacts and the nucleus, and implicates LPP in Scrib-associated functions (Petit, 2005).

Scrib family members interact armadillo repeat family proteins

Densin-180, a protein purified from the postsynaptic density fraction of the rat forebrain, is the founding member of a newly described family of proteins termed the LAP [leucine-rich repeats and PSD-95/Dlg-A/ZO-1 (PDZ) domains] family that plays essential roles in establishment of cell polarity. To identify Densin-180-binding proteins, a yeast two-hybrid library was screened using the carboxyl-terminal fragment of Densin-180 containing PDZ domain as bait, and delta-catenin/neural plakophilin-related armadillo repeat protein (NPRAP) was isolated as a Densin-180-interacting protein. delta-catenin/NPRAP, a member of the armadillo repeat family, is a nervous system-specific adherens junction protein originally discovered as an interactor with presenilin-1, a protein involved in Alzheimer's disease. Densin-180 PDZ domain binds the COOH terminus of delta-catenin/NPRAP containing the PDZ domain-binding sequence. Endogenous Densin-180 was co-immunoprecipitated with delta-catenin/NPRAP and N-cadherin. Although Densin-180 is reported to be a transmembrane protein, Densin-180 is not accessible to surface biotinylation in dissociated hippocampal neurons; hence Densin-180 may be a cytosolic protein. Densin-180 co-localizes with delta-catenin/NPRAP at synapses in delta-catenin/NPRAP and may be involved in organization of the synaptic cell-cell junction through interaction with the delta-catenin/NPRAP-N-cadherin complex (Izawa, 2002a).

ERBIN, an ErbB2 receptor-interacting protein, belongs to a recently described family of proteins termed the LAP [leucine-rich repeats and PSD-95/dLg-A/ZO-1 (PDZ) domains] family, which has essential roles in establishment of cell polarity. To identify new ERBIN-binding proteins, a yeast two-hybrid library was screened, using the carboxyl-terminal fragment of ERBIN containing PDZ domain as the bait, and p0071 (also called plakophilin-4) was isolated as an ERBIN-interacting protein. p0071 is a member of the p120 catenin family, defined as proteins with 10 armadillo repeats; these proteins localize along the cell-cell border. The ERBIN PDZ domain binds the COOH-terminus of p0071 containing the PDZ domain-binding sequence. Endogenous ERBIN co-immunoprecipitates with p0071. In fully polarized Madin-Darby canine kidney (MDCK) cells, ERBIN co-localizes largely with beta-catenin and partly with desmoplakin along the lateral plasma membrane domain. At these cell-cell contact regions, ERBIN co-localizes with p0071. Over-expression of the dominant active forms of Cdc42, Rac1 or RhoA, Rho family small GTPases, results in a marked accumulation of ERBIN at the cell-cell contacts of MDCK and HeLa cells. These results show that ERBIN interacts in vivo with p0071 and that it may be involved in the organization of adherens junctions and the desmosomes of epithelia. In addition, the subcellular localization of ERBIN might be regulated by Rho family small GTPases (Izawa, 2002b).

Integrity of epithelial tissues relies on the proper apical-basolateral polarity of epithelial cells. Members of the LAP (LRR and PDZ) protein family such as LET-413 and Scribble are involved in maintaining epithelial cell polarity in Caenorhabditis elegans and Drosophila melanogaster, respectively. Erbin is a mammalian LET-413 homologue interacting with ERBB2/HER2, an epidermal growth factor receptor family member. Erbin and ERBB2/HER2 are located in the basolateral membranes of epithelial cells. Erbin interacts with p0071 (also called plakophilin-4), an armadillo repeat protein linked to the cytoskeleton. Erbin binds to p0071 in vitro and in vivo in a PDZ domain-dependent manner, and both proteins colocalize in desmosomes of epithelial cells. Using a dominant negative approach, it was found that integrity of epithelial cell monolayer is impaired when interaction between Erbin and p0071 is disrupted. It is proposed that Erbin is connected by p0071 to cytoskeletal networks in an interaction crucial for epithelial homeostasis (Jaulin-Bastard, 2002).

Erbin is a member of the LAP (leucine-rich repeat and PDZ domain) protein family. A C-terminally displayed phage peptide library was used to identify optimal ligands for the Erbin PDZ domain. Phage-selected peptides were type 1 PDZ ligands that bind with high affinity and specificity to the Erbin PDZ domain in vitro. These peptides most closely resemble the C-terminal PDZ domain-binding motifs of three p120-related catenins: delta-catenin, ARVCF, and p0071 (DSWV-COOH). Analysis of the interactions of the Erbin PDZ domain with synthetic peptides matching the C termini of ARVCF or delta-catenin also demonstrates specific high affinity binding. The interactions between the Erbin PDZ domain and both ARVCF and delta-catenin were characterized in vitro and in vivo. The Erbin PDZ domain co-localizes and coprecipitates with ARVCF or delta-catenin complexed with beta-catenin and E/N-cadherin. Mutagenesis and peptide competition experiments show that the association of Erbin with the cadherin-catenin complex is mediated by the interaction of its PDZ domain with the C-terminal PDZ domain-binding motifs (DSWV-COOH) of ARVCF and delta-catenin. Finally, endogenous delta-catenin and Erbin were shown to co-localize in and co-immunoprecipitate from neurons. These results suggest that delta-catenin and ARVCF may function to mediate the association of Erbin with the junctional cadherin-catenin complex. They also demonstrate that C-terminal phage-display technology can be used to predict physiologically relevant ligands for PDZ domains (Laura, 2002).

Zebrafish neural tube morphogenesis requires Scribble-dependent oriented cell divisions

How control of subcellular events in single cells determines morphogenesis on the scale of the tissue is largely unresolved. The stereotyped cross-midline mitoses of progenitors in the zebrafish neural keel provide a unique experimental paradigm for defining the role and control of single-cell orientation for tissue-level morphogenesis in vivo. This study shows that the coordinated orientation of individual progenitor cell division in the neural keel is the cellular determinant required for morphogenesis into a neural tube epithelium with a single straight lumen. This study shows that Scribble is required for oriented cell division, and its function in this process is independent of canonical apicobasal and planar polarity pathways. A role is identified for Scribble in controlling clustering of β-catenin foci in dividing progenitors. Loss of either Scrib or N-cadherin results in abnormally oriented mitoses, reduced cross-midline cell divisions, and similar neural tube defects. It is proposed that Scribble-dependent nascent cell-cell adhesion clusters between neuroepithelial progenitors contribute to define orientation of their cell division. Finally, the data demonstrate that while oriented mitoses of individual cells determine neural tube architecture, the tissue can in turn feed back on its constituent cells to define their polarization and cell division orientation to ensure robust tissue morphogenesis (ŽZigman, 2011).

Dual roles of zygotic and maternal Scribble1 in neural migration and convergent extension movements in zebrafish embryos

In the developing vertebrate hindbrain, the characteristic trajectory of the facial (nVII) motor nerve is generated by caudal migration of the nVII motor neurons. The nVII motor neurons originate in rhombomere (r) 4, and migrate caudally into r6 to form the facial motor nucleus. Using a transgenic zebrafish line that expresses green fluorescent protein (GFP) in the cranial motor neurons, two novel mutants, designated landlocked (llk) and off-road (ord), have been isolated that both show highly specific defects in the caudal migration of the nVII motor neurons. The landlocked locus contains the gene scribble1 (scrb1), and its zygotic expression is required for migration of the nVII motor neurons mainly in a non cell-autonomous manner. Taking advantage of the viability of the llk mutant embryos, it was found that maternal expression of scrb1 is required for convergent extension (CE) movements during gastrulation. Furthermore, a genetic interaction is seen between scrb1 and trilobite(tri)/strabismus(stbm) in CE. The dual roles of the scrb1 gene in both neuronal migration and CE provide a novel insight into the underlying mechanisms of cell movement in vertebrate development (Wada, 2005).

Although the results suggest that there is a genetic interaction between scrb1 and stbm, it is not known whether the PDZ domains of Scrb directly interact with the PDZ-binding domain of Stbm. In Drosophila, the second PDZ domain of Scrb interacts with Dlg via GUKH (guanylate kinase holder protein) to form a scaffolding complex at synaptic junctions. Furthermore, Dlg interacts with Stbm and this complex is required for plasma membrane formation in epithelial cells. These results suggest that Scrb, Stbm and Dlg may constitute a functional complex during the formation of membrane structures. If Tri/Stbm and Llk/Scrb1 form a functional complex, this complex would probably have two sites that associate with membranes: the transmembrane domain of Tri/Stbm and the LRR domain of Llk/Scrb1. Knock-down of Tri/Stbm with overexpression of Llk/Scrb1 leads to the most severe impairment of CE. These results indicate that Tri/Stbm may be required for localization of Llk/Scrb1 protein to the specific site of the membrane where they are anchored and function together. Release of membrane-associated Llk/Scrb1 from such positional constraint in the absence of Stbm may have more markedly perturbed the functional protein complexes controlling CE than simple overexpression of Scrb1 in the presence of Stbm (Wada, 2005).

The Scrb1rw16 protein, which has a single amino acid substitution in the first PDZ domain, has lower activity than the wild-type protein to rescue migration of the nVII motor neurons in the llk mutation. Similarly, overexpression of Scrb1rw16 induces CE defects to a lesser extent than that of wild-type Scrb1 protein. These results indicate that the first PDZ domain is also essential for Scrb1 activity. The first PDZ domain of Llk/Scrb1 may interact with another, as yet unidentified, component to establish a multi-protein complex required for its function (Wada, 2005).

Mammalian Scribble polarity protein regulates epithelial cell adhesion and migration through E-cadherin

Scribble (Scrib) is a conserved polarity protein required in Drosophila for synaptic function, neuroblast differentiation, and epithelial polarization. It is also a tumor suppressor. In rodents, Scrib has been implicated in receptor recycling and planar polarity but not in apical/basal polarity. Knockdown of Scrib disrupts adhesion between Madin-Darby canine kidney epithelial cells. As a consequence, the cells acquire a mesenchymal appearance, migrate more rapidly, and lose directionality. Although tight junction assembly is delayed, confluent monolayers remain polarized. These effects are independent of Rac activation or Scrib binding to ßPIX. Rather, Scrib depletion disrupts E-cadherin-mediated cell-cell adhesion. The changes in morphology and migration are phenocopied by E-cadherin knockdown. Adhesion is partially rescued by expression of an E-cadherin-alpha-catenin fusion protein but not by E-cadherin-green fluorescent protein. These results suggest that Scrib stabilizes the coupling between E-cadherin and the catenins and are consistent with the idea that mammalian Scrib could behave as a tumor suppressor by regulating epithelial cell adhesion and migration (Qin, 2005).

A betaPIX-PAK2 complex confers protection against Scrib-dependent and cadherin-mediated apoptosis

During epithelial morphogenesis, a complex comprising the βPIX (PAK-interacting exchange factor β) and class I PAKs (p21-activated kinases) is recruited to adherens junctions. Scrib, the mammalian ortholog of the Drosophila polarity determinant and tumor suppressor Scribble, binds βPIX directly. Scrib is also targeted to adherens junctions by E-cadherin, where Scrib strengthens cadherin-mediated cell-cell adhesion. Although a role for the Scrib-βPIX-PAK signaling complex in promoting membrane protrusion at wound edges has been elucidated, a function for this complex at adherens junctions remains unknown. This study establish, in cultured mammalian cells, that Scrib targets βPIX and PAK2 to adherens junctions where a βPIX-PAK2 complex counterbalances apoptotic stimuli transduced by Scrib and elicited by cadherin-mediated cell-cell adhesion. Moreover, it was shown that this signaling pathway regulates cell survival in response to osmotic stress. Finally, it was determined that in suspension cultures, the Scrib-βPIX-PAK2 complex functions to regulate anoikis elicited by cadherin engagement, with Scrib promoting and the βPIX-PAK2 complex suppressing anoikis, respectively. These findings demonstrate that the Scrib-βPIX-PAK2 signaling complex functions as an essential modulator of cell survival when localized to adherens junctions of polarized epithelia. The activity of this complex at adherens junctions is thereby essential for normal epithelial morphogenesis and tolerance of physiological stress. Furthermore, when localized to adherens junctions, the Scrib-βPIX-PAK2 signaling complex serves as a key determinant of anoikis sensitivity, a pivotal mechanism in tumor suppression. Thus, this work also reveals the need to expand the definition of anoikis to include a central role for adherens junctions (Frank, 2012).

This study has established that differentiated epithelial cells rely on a survival signaling network associated with adherens junctions that is distinct from that used at cell-matrix adhesions. Specifically, it was demonstrated that epithelial cells are critically dependent on Scrib-mediated localization of a βPIX-PAK2 complex to adherens junctions to counterbalance the apoptosis-promoting effects of E-cadherin engagement. As a result, disruption of βPIX-PAK2 signaling results in a near complete loss of epithelial viability at confluent density. Consistent with an established role in phosphorylating cellular targets involved in apoptosis, a functional kinase domain is required for PAK2-dependent survival signaling. In subconfluent cultures, where the βPIX-PAK2 complex localizes to focal adhesions, the complex is dispensable for cell survival. Thus, as epithelial cells form cell-cell junctions and polarize, they become critically dependent on a prosurvival signal provided by the βPIX-PAK2 complex at adherens junctions (Frank, 2012).

To date, the only direct evidence for the function of PAKs at adherens junctions is in the regulation of cadherin adhesiveness and actin-dependent cell contractility. While invoked by prior findings, the current results demonstrate for the first time that Scrib via its association with βPIX promotes targeting of PAK2 to adherens junctions. In cultured human keratinocytes, PAK1 has been reported to augment cadherin adhesiveness in response to activated Rac. PAK1 and PAK2 have also been reported to enhance and suppress, respectively, loss of cell-cell contacts in response to hepatocyte growth factor. The current results suggest that neither PAK1 nor PAK2 play a major role in steady state junctional integrity in MDCK cells. However, because MDCK cells express both PAK1 and PAK2, the possibility cannot be excluded that they may function redundantly in regulation of E-cadherin adhesiveness and/or junctional remodeling (Frank, 2012).

βPIX and PAKs have been most extensively characterized for their role in focal adhesion dynamics, where they play a coordinated role in regulating turnover of these integrin attachment sites and promoting directional motility. PAK1 and PAK2 appear to have nonredundant functions in cell invasion and motility. Moreover, there is evidence to suggest that when localized to focal adhesions, the βPIX-PAK complex promotes mitogenic signaling and that redistribution of the complex from focal adhesions to adherens junctions contributes to the cessation of epithelial proliferation and establishment of contact inhibition. Scrib also shuttles between adherens junctions and protrusive membrane structures where it promotes βPIX-PAK complex-dependent cell motility. It will be interesting to determine whether Scrib will play a role in redistribution of the βPIX-PAK2 complex from focal adhesions to adherens junctions, which occurs as cells undergo contact inhibition. In brief, taken together with the present work, these studies suggest that PAKs have distinct roles when localized to focal adhesions and adherens junctions. When localized to focal adhesions, PAKs participate in signaling pathways that control proliferation and motility, whereas at adherens junctions PAKs regulate adhesiveness and survival. As such, the translocation of the βPIX-PAK complex from to lateral membranes upon formation of stable adherens junctions likely plays a fundamental role in the transition from motile and mitogenic states to a nonmotile and quiescent state (Frank, 2012).

E-cadherin is a potent inhibitor of multiple signaling pathways and plays a fundamental role in suppression of motility and proliferation upon establishment of cell-cell contact. The ability of Scrib to promote E-cadherin adhesiveness may underlie some of its tumor suppressive potential in mammalian cells. However, increasing evidence suggests that Scrib also plays E-cadherin-independent roles in the regulation of signaling pathways, such as inhibition of ERK and AKT, as well as activation of Hippo signaling. These Scrib-dependent effects would be predicted to sensitize cells to apoptosis; a prediction supported by the present results, which furthermore establish that Scrib-mediated apoptosis is counterbalanced by its recruitment of active PAK2. Taken together, these findings demonstrate that Scrib transduces both pro- and anti-apoptotic stimuli (Frank, 2012).

Metastasis requires that cells tolerate the loss of matrix adhesion, i.e., that they are protected against anoikis. However, in spite of seminal work suggesting a role for cell-cell adhesion in modulating anoikis, the role for epithelial architecture in anoikis remains unappreciated and poorly characterized. Nevertheless, there is growing evidence indicating that loss of E-cadherin function suffices to abrogate anoikis. The current results support a key role of adherens junctions in modulating anoikis. In simple polarized epithelia, which likely rely on the summation of survival signals emanating from both focal adhesions and adherens junctions, apoptotic stimuli emanating from E-cadherin will sensitize cells to anoikis. In contrast, suprabasal cells in stratified epithelia depend exclusively on survival signals from adherens junctions. PAK2 is highly expressed in suprabasal keratinocytes, which may suggest that upregulation of PAK2-dependent survival signals emanating from adherens junctions are required to offset loss of integrin-mediated survival signaling. In summary, the current results indicate that E-cadherin does not function simply as a prosurvival or proapoptotic factor but rather as a regulatory node to coordinate death and survival signaling. The balance between pro- and antiapoptotic signaling emanating from adherens junctions plays a key role in epithelial cell viability and is likely essential in regulating diverse processes, including epithelial morphogenesis, wound healing, physiological stress, and metastasis (Frank, 2012).

Non-canonical Wnt signaling regulates cell polarity in female reproductive tract development via van gogh-like 2

Wnt signaling effectors direct the development and adult remodeling of the female reproductive tract (FRT); however, the role of non-canonical Wnt signaling has not been explored in this tissue. The non-canonical Wnt signaling protein van gogh-like 2 is mutated in loop-tail (Lp) mutant mice (Vangl2Lp), which display defects in multiple tissues. Vangl2Lp mutant uterine epithelium displays altered cell polarity, concommitant with changes in cytoskeletal actin and scribble (scribbled, Scrb1) localization. The postnatal mutant phenotype is an exacerbation of that seen at birth, exhibiting more smooth muscle and reduced stromal mesenchyme. These data suggest that early changes in cell polarity have lasting consequences for FRT development. Furthermore, Vangl2 is required to restrict Scrb1 protein to the basolateral epithelial membrane in the neonatal uterus, and an accumulation of fibrillar-like structures observed by electron microscopy in Vangl2Lp mutant epithelium suggests that mislocalization of Scrb1 in mutants alters the composition of the apical face of the epithelium. Heterozygous and homozygous Vangl2Lp mutant postnatal tissues exhibit similar phenotypes and polarity defects and display a 50% reduction in Wnt7a levels, suggesting that the Vangl2Lp mutation acts dominantly in the FRT. These studies demonstrate that the establishment and maintenance of cell polarity through non-canonical Wnt signaling are required for FRT development (Vandenberg, 2009).

Planar polarity pathway and Nance-Horan syndrome-like 1b have essential cell-autonomous functions in neuronal migration

Components of the planar cell polarity (PCP) pathway are required for the caudal tangential migration of facial branchiomotor (FBM) neurons, but how PCP signaling regulates this migration is not understood. In a forward genetic screen, a new gene was identified, nhsl1b, that is required for FBM neuron migration. nhsl1b encodes a WAVE-homology domain-containing protein related to human Nance-Horan syndrome (NHS) protein and Drosophila GUK-holder (Gukh), which have been shown to interact with components of the WAVE regulatory complex that controls cytoskeletal dynamics and with the polarity protein Scribble, respectively. Nhsl1b localizes to FBM neuron membrane protrusions and interacts physically and genetically with Scrib to control FBM neuron migration. Using chimeric analysis, it was shown that FBM neurons have two modes of migration: one involving interactions between the neurons and their planar-polarized environment, and an alternative, collective mode involving interactions between the neurons themselves. The first mode of migration requires the cell-autonomous functions of Nhsl1b and the PCP components Scrib and Vangl2 in addition to the non-autonomous functions of Scrib and Vangl2, which serve to polarize the epithelial cells in the environment of the migrating neurons. These results define a role for Nhsl1b as a neuronal effector of PCP signaling and indicate that proper FBM neuron migration is directly controlled by PCP signaling between the epithelium and the migrating neurons (Walsh, 2011).

Scribble1/AP2 complex coordinates NMDA receptor endocytic recycling

The appropriate trafficking of glutamate receptors to synapses is crucial for basic synaptic function and synaptic plasticity. It is now accepted that NMDA receptors (NMDARs; see Drosophila NMDA receptors) internalize and are recycled at the plasma membrane but also exchange between synaptic and extrasynaptic pools; these NMDAR properties are also key to governing synaptic plasticity. Scribble1 is a large PDZ protein required for synaptogenesis and synaptic plasticity. This study shows that the level of Scribble1 is regulated in an activity-dependent manner and that Scribble1 controls the number of NMDARs at the plasma membrane. Notably, Scribble1 prevents GluN2A subunits from undergoing lysosomal trafficking and degradation by increasing their recycling to the plasma membrane following NMDAR activation. Finally, it was shown that a specific YxxR motif on Scribble1 controls these mechanisms through a direct interaction with AP2 (see Drosophila AP2). Altogether, these findings define a molecular mechanism to control the levels of synaptic NMDARs via Scribble1 complex signaling (Piguel, 2014).

Scribble, APC and cell cycle

Human scribble (hScrib), human homolog of the Drosophila tumor suppressor Scribble, has been identified a substrate of human papillomavirus E6 oncoproteins for ubiquitin-mediated degradation dependent on ubiquitin-protein ligase E6AP. Human Scribble, classified as a LAP protein containing leucine-rich repeats and PDZ domains, interacts with E6 through its PDZ domains and C-terminal PDZ domain-binding motif of E6 protein. Interaction between human Discs Large (hDlg), a substrate of E6 for the ubiquitin-mediated degradation, and adenomatous polyposis coli (APC) has been shown. This study investigated whether hScrib and APC interact with each other in vitro and in vivo. Interaction between hScrib and APC is mediated by the PDZ domains 1 and 4 of hScrib and C-terminal PDZ domain-binding motif of APC. Human Scribble co-localizes with APC at the synaptic sites of hippocampal neuron and at the tip of membrane protrusion in the epithelial cell line. Interference of the interaction between hScrib and APC causes disruption of adherens junction. Knockdown of hScrib expression by RNAi disrupts localization of APC at the adherens junction. These data suggest that hScrib may participate in the hDlg-APC complex through its PDZ domains and regulate cell cycle and neural function by associating with APC (Takizawa, 2006).

Drosophila tumor suppressor Scribble has been identified as an apical-basolateral polarity determinant in epithelia. A human homolog of Drosophila Scribble, human Scribble (hScrib), has been identified as a protein targeted by human papillomavirus E6 for the ubiquitin-mediated degradation dependent on E6AP, a cellular ubiquitin-protein ligase. Human Scribble is classified as a LAP protein, having leucine-rich repeats (LRRs) and PDZ domains. Whether hScrib, which is thought to have a role in polarity determination based on the data of its Drosophila homolog, is involved in cell-cycle regulation and proliferation control of epithelia was investigated. Transfection of hScrib inhibits cell-cycle progression from G1 to S phase, and it up- and down-regulates expression of adenomatous polyposis coli and cyclins A and D1, respectively. Knockdown of hScrib expression by siRNA leads to cell-cycle progression from G1 to S phase. Functional domain mapping was explored to reveal which domains of hScrib are critical for its cellular proliferation control and localization at the basolateral membrane. LRRs and PDZ domain 1 were found to be indispensable for hScrib to inhibit cell growth by blocking cell-cycle progression and to keep its proper localization. These data indicate that basolateral membrane localization of hScrib is closely related to its proliferation control. These findings suggest the possibility that hScrib is involved in signal transduction to negatively regulate cell proliferation by localizing at the basolateral membrane of epithelial cells through LRRs and PDZ domains (Nagasaka, 2006).

Scrib is required for epithelial cell identity and prevents epithelial to mesenchymal transition in the mouse

This study asked if the mouse homolog of Drosophila Scribbled is required for establishment and/or maintenance of epithelial identity in vivo. To do so, Scrib was conditionally deleted in the head ectoderm tissue that gives rise to both the ocular lens and the corneal epithelium. Deletion of Scrib in the lens resulted in a change in epithelial cell shape from cuboidal to flattened and elongated. Early in the process, the cell adhesion protein, E-cadherin, and apical polarity protein, ZO-1, were downregulated and the myofibroblast protein, αSMA, was upregulated, suggesting epithelial-mesenchymal transition (EMT) was occurring in the Scrib deficient lenses. Correlating temporally with the upregulation of alphaSMA, Smad3 and Smad4, TGFβ signaling intermediates, accumulated in the nucleus and Snail, a TGFβ target and transcriptional repressor of the gene encoding E-cadherin, was upregulated. Pax6, a lens epithelial transcription factor required to maintain lens epithelial cell identity also was downregulated. Loss of Scrib in the corneal epithelium also led to molecular changes consistent with EMT, suggesting that the effect of Scrib deficiency was not unique to the lens. Together, these data indicate that mammalian Scrib is required to maintain epithelial identity and that loss of Scrib can culminate in EMT, mediated, at least in part, through TGFβ signaling (Yamben, 2013).

Mislocalization of the cell polarity protein Scribble promotes mammary tumorigenesis and is associated with basal breast cancer

Scribble (SCRIB) localizes to cell-cell junctions and regulates establishment of epithelial cell polarity. Loss of expression of SCRIB functions as a tumor suppressor in Drosophila and mammals, conversely, overexpression of SCRIB promotes epithelial differentiation in mammals. This study reports that SCRIB is frequently amplified, mRNA over-expressed and protein is mislocalized from cell-cell junctions in human breast cancers. High levels of SCRIB mRNA are associated with poor clinical prognosis identifying an unexpected role for SCRIB in breast cancer. Transgenic mice expressing a SCRIB mutant (Pro 305 to Leu (P305L)) that fails to localize to cell-cell junctions, under the control of the mouse mammary tumor virus long terminal repeat promoter, develop multifocal hyperplasia that progresses to highly pleomorphic and poorly differentiated tumors with basal characteristics. SCRIB interacts with PTEN and the expression of P305L, but not wild-type SCRIB, promotes an increase in PTEN levels in the cytosol. Overexpression of P305L, but not wild type SCRIB, activates the Akt/mTOR/S6K signaling pathway. Human breast tumors overexpressing SCRIB have high levels of S6K but do not harbor mutations in PTEN or PIK3CA, identifying SCRIB amplification as a mechanism of activating PI3K signaling in tumors without mutations in PIK3CA or PTEN. Thus, this study has demonstrated that high levels of mislocalized SCRIB functions as a neomorph to promote mammary tumorigenesis by affecting subcellular localization of PTEN and activating an Akt/mTOR/S6kinase signaling pathway (Feigin, 2014).

The cell fate determinant Scribble is required for maintenance of hematopoietic stem cell function

Cell fate determinants influence self-renewal potential of hematopoietic stem cells, Scribble and Llgl1 belong to the Scribble polarity complex and reveal tumor-suppressor function in Drosophila. In hematopoietic cells, genetic inactivation of Llgl1 leads to expansion of the stem cell pool and increases self-renewal capacity without conferring malignant transformation. This study shows that that genetic inactivation of its putative complex partner Scribble results in functional impairment of hematopoietic stem cells (HSC) over serial transplantation and during stress. Although loss of Scribble deregulates transcriptional downstream effectors involved in stem cell proliferation, cell signaling, and cell motility, these effectors do not overlap with transcriptional targets of Llgl1. Binding partner analysis of Scribble in hematopoietic cells using affinity purification followed by mass spectometry confirms its role in cell signaling and motility but not for binding to polarity modules described in drosophila. Finally, requirement of Scribble for self-renewal capacity also affects leukemia stem cell function. Thus, Scribble is a regulator of adult HSCs, essential for maintenance of HSCs during phases of cell stress (Mohr, 2018).

Scribble, Erbin, and Lano redundantly regulate epithelial polarity and apical adhesion complex

The basolateral protein Scribble (Scrib), a member of the LAP protein family, is essential for epithelial apicobasal polarity (ABP) in Drosophila. However, a conserved function for this protein in mammals is unclear. This study showed that the crucial role for Scrib in ABP has remained obscure due to the compensatory function of two other LAP proteins, Erbin and Lano. A combined Scrib/Erbin/Lano knockout disorganizes the cell-cell junctions and the cytoskeleton. It also results in mislocalization of several apical (Par6, aPKC, and Pals1) and basolateral (Llgl1 and Llgl2) identity proteins. These defects can be rescued by the conserved "LU" region of these LAP proteins. Structure-function analysis of this region determined that the so-called LAPSDb domain is essential for basolateral targeting of these proteins, while the LAPSDa domain is essential for supporting the membrane basolateral identity and binding to Llgl. In contrast to the key role in Drosophila, mislocalization of Llgl proteins does not appear to be critical in the scrib ABP phenotype (Choi, 2019).

An interaction between Scribble and the NADPH oxidase complex controls M1 macrophage polarization and function

The polarity protein Scribble (SCRIB) regulates apical-basal polarity, directional migration and tumour suppression in Drosophila and mammals. This study reports that SCRIB is an important regulator of myeloid cell functions including bacterial infection and inflammation. SCRIB interacts directly with the NADPH oxidase (NOX; see Drosophila Nox) complex in a PSD95/Dlg/ZO-1 (PDZ)-domain-dependent manner and is required for NOX-induced reactive oxygen species (ROS) generation in culture and in vivo. On bacterial infection, SCRIB localized to phagosomes in a leucine-rich repeat-dependent manner and promoted ROS production within phagosomes to kill bacteria. Unexpectedly, SCRIB loss promoted M1 macrophage polarization and inflammation. Thus, SCRIB uncouples ROS-dependent bacterial killing activity from M1 polarization and inflammatory functions of macrophages. Modulating the SCRIB-NOX pathway can therefore identify ways to manage infection and inflammation with implications for chronic inflammatory diseases, sepsis and cancer (Zheng, 2016).

Downregulation of SCRIB expression in a macrophage cell line, RAW 264.7, significantly impaired phorbol 12-myristate 13-acetate (PMA)-induced generation of ROS. In addition, primary macrophages were used from an inducible SCRIB knockdown mouse model where SCRIB short hairpin RNA (shRNA) expression was controlled by a tetracycline-response element (TRE) at the COLA1 locus (SCRIB ishRNA). Reverse tetracycline transactivator (rtTA) expression from a ROSA26 promoter facilitated doxycycline (Dox)-inducible expression of SCRIB ishRNA and GFP in all cells, including isolated bone-marrow-derived macrophages (BMMs). Inducible loss of SCRIB did not affect BMM differentiation as monitored by expression of CD11b/F4/80 and cell morphology. Both PMA and lipopolysaccharide (LPS) induced ROS in wild-type cells and in SCRIB ishRNA cells in the absence of Dox stimulation. Dox-induced knockdown of SCRIB significantly impaired PMA- or LPS-induced ROS. In addition, PMA-induced ROS was significantly low in primary neutrophils from Dox-treated mice, demonstrating a role for SCRIB during ROS generation in primary myeloid cells (Zheng, 2016).

Apart from myeloid cells, in mouse embryo fibroblasts (MEFs) derived from SCRIB ishRNA mice platelet-derived growth factor (PDGF)-induced generation of ROS and tyrosine phosphorylation of the PDGF receptor were impaired, demonstrating a role for SCRIB in regulating ROS generation in multiple contexts. The decrease in PDGFR tyrosine phosphorylation is consistent with the well-established role for ROS in inactivating tyrosine phosphatase activity and promoting tyrosine phosphorylation (Zheng, 2016).

To determine the in vivo significance, SCRIB ishRNA mice were injected with LPS intraperitoneally and ROS was monitored in real time using L-012, a luminol-based chemiluminescence probe used for detecting NADPH oxidase-derived superoxide. LPS challenge induced a dramatic and sustained increase in ROS levels in -Dox, but not in +Dox mice in vivo. The lack of LPS-induced ROS in vivo was not due to defective immune biology because cells from the thymus, spleen and bone marrow of wild-type (WT) (+Dox and -Dox) and SCRIB ishRNA (+Dox and -Dox) mice showed comparable expression of differentiation markers on dendritic, neutrophil, monocyte or B and T cells (Zheng, 2016).

Macrophages generate ROS (superoxide (O2-)) using the NOX complex. LPS, PMA and PDGF induce ROS by activating the NOX protein complex (Park, 2004; Segal, 1993). NOX-mediated generation of ROS requires activation of Rac GTPases facilitated by the GTP exchange factor β-PIX13. Since β-PIX binds directly with SCRIB, LPS-induced Rac activation was analysed in SCRIB shRNA cells. Interestingly, LPS-induced Rac.GTP levels were twofold lower in SCRIB shRNA cells compared with Luc shRNA cells (Zheng, 2016).

Tests were performed to see whether SCRIB functions as a scaffold to recruit β-PIX to the NOX complex. β-PIX co-immunoprecipitated both p22phox (a component of the NOX complex) and SCRIB in Flag-taggedβ-PIX-expressing control cells, but not in RAW 264.7 SCRIB shRNA cells. The interaction was observed both before and after PMA stimulation, with a modest increase in association post PMA stimulation (Zheng, 2016).

SCRIB is a PSD95/Dlg/ZO-1 (PDZ)-domain-containing protein and PDZ domains frequently use carboxy-terminal residues to interact with its partners. Analysis of the carboxy-terminal 10 amino acids of all members of the NOX complex showed that only the C-terminal sequence of p22phox (192-DEVV-195) conforms to a class III PDZ-binding motif (X(unspecified)-D/E-X-Ψ(hydrophobic)). Amino-terminal Flag-tagged WT p22phox, but not the p22phox lacking the C-terminal 10 amino acids (p22Δ), interacted with T7 epitope-tagged SCRIB. p22phox is a common subunit present in all of the NOX complexes (NOX1-4); accordingly, SCRIB co-immunoprecipitated with NOXO1 (a component of NOX1 and 3) and Tks5 (a component of NOX4), identifying SCRIB as a member of all NOX complexes. Both the NOX1 and NOX3 complexes require Rac.GTP for their activity, supporting a role for SCRIB in these complexes (Zheng, 2016).

Endogenous p22phox from both RAW 264.7 and primary BMMs was able to co-immunoprecipitate SCRIB. Conversely, endogenous SCRIB co-immunoprecipitated p22phox. In contrast to SCRIB-p22phox association, interaction with other components of the NOX complex, gp91phox, p67phox and p40phox, was induced on LPS stimulation in a SCRIB-dependent manner. This is consistent with previous observations that LPS stimulation induces assembly of an active NOX complex (Zheng, 2016).

To determine whether SCRIB and p22phox interact directly with each other, bacterially produced 6xHis-tag fusions were used of each of the four PDZ domains (SCRIB-PDZ1, SCRIB-PDZ2, SCRIB-PDZ3 and SCRIB-PDZ4) and GST-tag-fused p22phox C-terminal tail (186-NPIPVTDEVV-195) (p22tail). Purified SCRIB-PDZ4, but not the other PDZ domains, bound directly to purified p22tail. Next, a nuclear magnetic resonance (NMR) spectroscopy experiment was performed on 15N-labelled SCRIB-PDZ4 with unlabelled p22tail peptide (residues 186-195) to identify the binding interface. By increasing the concentration of the peptide, nine peaks were observed displaying marked peak shifts (>0.2 normalized chemical shift change) in the 1H-15N heteronuclear single-quantum coherence spectra. These perturbed residues (Leu1111, Gly1112, Ile1113, Ser1114, Ile1115, Ala1144, Ala1145, Arg1178 and Val1180) were mapped onto the previously reported three-dimensional structure of unligated SCRIB-PDZ4 (PDB: 1UJU). In contrast, a p22phox peptide lacking the carboxy tail (residues 131-185) displayed no change in the NMR spectra of SCRIB-PDZ4. A typical PDZ domain consists of six β-strands (βA-βF) and two α-helices (αA and αB). The most affected residues Leu1111 and Gly1112 are located in the well-known target-carboxylate binding loop (1110-RLGI-1113), while other perturbed residues, Ser1114 and Ile1115, reside at the βB strand and Arg1178 is located within the αB helix. The identified p22tail binding site on SCRIB-PDZ4 coincides well with the canonical target-binding pocket found in many PDZ proteins. Accordingly a p22tail-docking model structure of SCRIB-PDZ4 was generated using the structure of the Erbin PDZ domain-ErbB2 peptide complex (PDB 1MFG18) as a template. Additional tests confirmed that the PDZ4 domain of SCRIB is essential for interaction with the NOX complex and for the SCRIB-dependent production of ROS (Zheng, 2016).

NOX complex-generated respiratory burst within phagosomes is required to kill invading bacteria. To investigate whether loss of SCRIB affects the ability of phagocytes to clear bacterial infection, SCRIB ishRNA mice were challenged with an intraperitoneal injection of Staphylococcus aureus. SCRIB ishRNA - Dox mice were fourfold more effective in clearing S. aureus from both the peritoneal cavity and the lung compared with mice on a Dox diet. To rule out a role for differences in neutrophil migration, purified neutrophils and blood preparations from SCRIB ishRNA + Dox or -Dox mice were analyzed in culture; myeloid cells from +Dox mice have a fourfold decrease in S. aureus killing compared with cells from SCRIB ishRNA - Dox or WT mice, demonstrating a cell-intrinsic defect. Furthermore, both RAW 264.7 control and SCRIB shRNA cells were equally competent in internalizing pHrodo dye-conjugated S. aureus particles demonstrating that bacterial phagocytosis was unaffected by loss of SCRIB (Zheng, 2016).

Next, the relationship between SCRIB and phagosomes was investigated. Structured illumination microscopy of RAW 264.7 cells 1.0 h post S. aureus infection demonstrated co-localization of SCRIB and p22phox around S. aureus. Both the PDZ4 mutant and WT SCRIB, expressed in the background of SCRIB shRNA, were effective in localizing to the bacteria, demonstrating that SCRIB-p22phox interaction was not required for SCRIB to localize to phagosomes. In both SCRIB WT and PDZ4 rescue cell lines, SCRIB was localized at a comparable distance to bacteria (median distance between 1.5-2.0 µm), as monitored by conventional confocal image analysis. In addition to RAW 264.7 cells, SCRIB accumulated near bacteria containing phagosomes in a human macrophage cell line Thp1 expressing RFP-SCRIB and infected with FITC-S. aureus demonstrating evolutionary conservation of the mechanism (Zheng, 2016).

In both Drosophila and mammalian epithelial cells, SCRIB regulates cell polarity in a membrane localization-dependent manner. A SCRIB mutant that fails to localize to the cell membrane, Pro305 to leucine (P305L), also failed to localize near bacteria, identifying a role for membrane localization in localizing SCRIB to phagosomes. Neither PDZ4 nor SCRIBP305L expression in RAW 264.7 SCRIB shRNA cells rescued PMA-induced generation of ROS, suggesting that both interaction with p22phox and localization to the phagosome were required for ROS generation (Zheng, 2016).

Generation of ROS within the phagosome is required for the ability of macrophages to kill bacteria. RAW 264.7 Luc shRNA and SCRIB shRNA cells were infected with S. aureus and incubated with cerium chloride, which reacts with ROS in phagosomes to form an electron-dense precipitate visible by transmission electron microscopy. Electron-dense precipitates of cerium ions were found around the S. aureus-containing phagosomes in shLuc cells, but not SCRIB shRNA cells, identifying an unexpected role for SCRIB as a regulator of ROS generation within phagocytic structures (Zheng, 2016).

Whether SCRIB regulates macrophage polarization was investigated because M1 macrophages are key regulators of host defence to pathogen infection and inflammation. RAW 264.7 and primary, bone marrow-derived, cells were stimulated with a combination of interferon gamma and LPS to induce M1 or IL-4 to induce M2 macrophage polarization. Surprisingly, cells lacking SCRIB (+Dox) showed a significant increase in the tendency to polarize towards the M1 lineage, as monitored by expression of IL-1β and Il-12β messenger RNA, but a decrease in the tendency to polarize towards the M2 lineage, as monitored by Fizz-1 mRNA expression. This phenotype was rescued by re-expression of SCRIB WT, but not by expression of PDZ4 or P305L mutants, demonstrating the need for p22phox interaction and membrane localization. Thus, the impaired bacterial killing in mice lacking SCRIB is not due to a defect in M1 polarization (Zheng, 2016).

To determine whether the hyper-inflammatory response relates to enhanced cytokine production, BMMs from SCRIB ishRNA mice were cultured, stimulated with LPS for 2.0 h and assayed for mRNA expression of inflammatory cytokines using quantitative PCR arrays. Pro-inflammatory cytokines were upregulated in SCRIB ishRNA + Dox compared with SCRIB ishRNA - Dox cells (2-10-fold) on LPS challenge. Consistent with the increase in transcript levels, protein levels of the major inflammatory cytokines, IL-6 and TNF (also known as TNFα), were significantly higher in SCRIB ishRNA + Dox mice compared with -Dox conditions. Inflammatory cytokines are primarily induced by the NF-κB transcription complex. However, neither the phosphorylation of IkB-alpha nor nuclear localization of p65 was altered by SCRIB loss. ROS can oxidize Cys62 of the p50 subunit of NF-κB ROS and interfere with its DNA-binding ability. Thus, the low levels of LPS-induced ROS in SCRIB knockdown cells can paradoxically increase the DNA-binding capacity of NF-κB to DNA and promote expression of inflammatory cytokines. Consistent with this possibility, the p65 subunit of the NF-κB complex was bound at significantly higher levels on both the IL-6 and TNF promoter in cells lacking SCRIB as determined by chromatin immunoprecipitation (ChIP). However, polymerase II occupancy on these promoters was not affected by loss of SCRIB. In addition to macrophages, dendritic cells are also sensitive to differences in ROS levels. Dendritic cells with low levels of ROS show a heightened response to LPS challenge compared with those with high levels of ROS28. Bone-marrow-derived dendritic cells (BMDCs) showed an elevated expression of canonical activation markers, such as CD80, CD86 and MHCII, in the absence of SCRIB expression (+Dox) and with LPS stimulation. Thus, loss of SCRIB results in increased DC sensitivity to LPS stimulation, which potentially exacerbates the hyper-inflammatory phenotype and lethal response observed with LPS challenge of ishSCRIB + Dox mice (Zheng, 2016).

In addition to its role in the clearance of bacterial infections, NADPH oxidase is also an important regulator of inflammation. Individuals with chronic granulomatous disease, caused by mutations in members of the NOX complex, show hyper-inflammatory symptoms such as development of granuloma, Crohn's-like disease and pulmonary fibrosis. p47phox knockout mice, in addition to having a defect in clearing bacterial infection due to decreased generation of ROS, show a hyper-inflammatory response on LPS challenge. The current observations are thus consistent with the phenotypes observed in patients with a defective NOX complex and those reported for mouse models with mutations in the NOX complex, providing genetic support to the conclusion that SCRIB is a member of the NOX complex. In addition, unexpected finding is reported that SCRIB is a regulator of M1/M2 polarization. Thus, this study defines a function for SCRIB in cells of the myeloid lineage. Apart from myeloid cells, the NOX complex plays an important role in the central nervous system including microglia function, astrocyte survival, neuronal polarization, axonal growth and neurodegenerative disease. Thus, a better understanding of the SCRIB-NOX pathway can provide insights into neurological diseases, managing bacterial infection, modulating inflammation and tumour cell killing with implications for chronic inflammatory diseases, sepsis and cancer (Zheng, 2016).



REFERENCES

Search PubMed for articles about Drosophila scribbled

Albertson, R., and Doe, C. Q. (2003). Dlg, Scrib, and Lgl regulate neuroblast cell size and mitotic spindle asymmetry. Nat. Cell Biol. 5: 166-170. 12545176

Albertson, R., Chabu, C., Sheehan, A. and Doe, C. Q. (2004). Scribble protein domain mapping reveals a multistep localization mechanism and domains necessary for establishing cortical polarity. J. Cell Sci. 117: 6061-6070. 15536119

Andersen, D. S., Colombani, J., Palmerini, V., Chakrabandhu, K., Boone, E., Rothlisberger, M., Toggweiler, J., Basler, K., Mapelli, M., Hueber, A. O. and Leopold, P. (2015). The Drosophila TNF receptor Grindelwald couples loss of cell polarity and neoplastic growth. Nature. PubMed ID: 25874673

Apperson, M. L., Moon, I. S. and Kennedy, M. B. (1996). Characterization of densin-180, a new brain-specific synaptic protein of the O-sialoglycoprotein family. J. Neurosci. 16: 6839-52. PubMed Citation: 8824323

Audebert, S., et al. (2004). Mammalian Scribble forms a tight complex with the betaPIX exchange factor. Curr. Biol. 14(11): 987-95. 15182672

Bahri, S., et al. (2010). The leading edge during dorsal closure as a model for epithelial plasticity: Pak is required for recruitment of the Scribble complex and septate junction formation. Development 137(12): 2023-32. PubMed Citation: 20501591

Biederer, T. and Sudhof, T. C. (2000). Mints as adaptors. Direct binding to neurexins and recruitment of munc18. J Biol Chem 275(51): 39803-39806. PubMed ID: 11036064

Bilder, D. and Perrimon, N. (2000a). Localization of apical epithelial determinants by the basolateral PDZ protein Scribble. Nature 403: 676-680. PubMed Citation: 10688207

Bilder, D., Li, M. and Perrimon, N. (2000b). Cooperative regulation of cell polarity and growth by Drosophila tumor suppressors. Science 289(5476): 113-116. PubMed Citation: 10884224

Bilder, D., et al. (2000c). Collective nomenclature for LAP proteins. Nat. Cell Biol. 2: E114. PubMed Citation: 10878817

Bilder, D., Schober, M. and Perrimon, N. (2003). Integrated activity of PDZ protein complexes regulates epithelial polarity. Nat. Cell Biol. 5(1): 53-8. 12510194

Bonello, T. T., Choi, W. and Peifer, M. (2019). Scribble and Discs-large direct initial assembly and positioning of adherens junctions during the establishment of apical-basal polarity. Development 146(22). PubMed ID: 31628110

Borg, J. P., et al. (2000). ERBIN: a basolateral PDZ protein that interacts with the mammalian ERBB2/HER2 receptor. Nat. Cell Biol. 2: 407-414. PubMed Citation: 10878805

Bossinger, O., et al. (2001). Zonula adherens formation in Caenorhabditis elegans requires dlg-1, the homologue of the Drosophila gene discs large. Dev. Bio. 230: 29-42. 11161560

Brumby, A. M. and Richardson, H. E. (2003). scribble mutants cooperate with oncogenic Ras or Notch to cause neoplastic overgrowth in Drosophila. EMBO J. 22: 5769-5779. 14592975

Brumby, A., Secombe, J., Horsfield, J., Coombe, M., Amin, N., Coates, D., Saint, R. and Richardson, H. (2004). A genetic screen for dominant modifiers of a cyclin E hypomorphic mutation identifies novel regulators of S-phase entry in Drosophila. Genetics 168(1): 227-51. 15454540

Bunker, B.D., Nellimoottil, T.T., Boileau, R.M., Classen, A.K. and Bilder, D. (2015). The transcriptional response to tumorigenic polarity loss in Drosophila. Elife [Epub ahead of print]. PubMed ID: 25719210

Carvalho, C.A., Moreira, S., Ventura, G., Sunkel, C.E. and Morais-de-Sá, E. (2015). Aurora A triggers Lgl cortical release during symmetric division to control planar spindle orientation. Curr Biol. 25: 53-60. PubMed ID: 25484294

Chen, C. L., Schroeder, M. C., Kango-Singh, M., Tao, C. and Halder, G. (2012). Tumor suppression by cell competition through regulation of the Hippo pathway. Proc. Natl. Acad. Sci. 109(2): 484-9. PubMed Citation: 22190496

Choi, J., Troyanovsky, R. B., Indra, I., Mitchell, B. J. and Troyanovsky, S. M. (2019). Scribble, Erbin, and Lano redundantly regulate epithelial polarity and apical adhesion complex. J Cell Biol. PubMed ID: 31147384

de Wit, J., Sylwestrak, E., O'Sullivan, M. L., Otto, S., Tiglio, K., Savas, J. N., Yates, J. R., 3rd, Comoletti, D., Taylor, P. and Ghosh, A. (2009). LRRTM2 interacts with Neurexin1 and regulates excitatory synapse formation. Neuron 64(6): 799-806. PubMed ID: 20064388

Doggett, K., Turkel, N., Willoughby, L. F., Ellul, J., Murray, M. J., Richardson, H. E. and Brumby, A. M. (2015). BTB-zinc finger oncogenes are required for Ras and Notch-driven tumorigenesis in Drosophila. PLoS One 10: e0132987. PubMed ID: 26207831

Dow, L. E., et al. (2003). hScrib is a functional homologue of the Drosophila tumour suppressor Scribble. Oncogene 22(58): 9225-30. 14681682

Feigin, M. E., Akshinthala, S. D., Araki, K., Rosenberg, A. Z., Muthuswamy, L. B., Martin, B., Lehmann, B. D., Berman, H. K., Pietenpol, J. A., Cardiff, R. D. and Muthuswamy, S. K. (2014). Mislocalization of the cell polarity protein Scribble promotes mammary tumorigenesis and is associated with basal breast cancer. Cancer Res [Epub ahead of print]. PubMed ID: 24662921

Frank, S. R., Bell, J. H., Frodin, M. and Hansen, S. H. (2012). A betaPIX-PAK2 complex confers protection against Scrib-dependent and cadherin-mediated apoptosis. Curr Biol 22: 1747-1754. PubMed ID: 22863318

Fujita, Y., et al. (1998). Tomosyn: a syntaxin-1-binding protein that forms a novel complex in the neurotransmitter release process. Neuron 20: 905-15. PubMed Citation: 9620695

Ganguly, I., Mackay, T. F. and Anholt, R. R. (2003). Scribble is essential for olfactory behavior in Drosophila melanogaster. Genetics 164(4): 1447-57. 12930751

Gui, J., Huang, Y. and Shimmi, O. (2016). Scribbled optimizes BMP signaling through its receptor internalization to the Rab5 endosome and promote robust epithelial morphogenesis. PLoS Genet 12: e1006424. PubMed ID: 27814354

Igaki, T., et al. (2006). Loss of cell polarity drives tumor growth and invasion through JNK activation in Drosophila. Curr. Biol. 16: 1139-1146. 16753569

Igaki, T., et al. (2009). Intrinsic tumor suppression and epithelial maintenance by endocytic activation of Eiger/TNF signaling in Drosophila. Dev. Cell 16: 458-465. PubMed Citation: 19289090

Izawa, I., Nishizawa, M., Ohtakara, K. and Inagaki, M. (2002a). Densin-180 interacts with delta-catenin/neural plakophilin-related armadillo repeat protein at synapses. J. Biol. Chem. 277: 5345-5350. 11729199

Izawa, I., Nishizawa, M., Tomono, Y., Ohtakara, K., Takahashi, T. and Inagaki, M. (2002b). ERBIN associates with p0071, an armadillo protein, at cell-cell junctions of epithelial cells. Genes Cells 7: 475-485. 12047349

Jaulin-Bastard, F., Arsanto, J. P., le Bivic, A., Navarro, C., Vely, F., Saito, H., Marchetto, S., Hatzfeld, M., Santoni, M. J., Birnbaum, D., et al. (2002). Interaction between Erbin and a Catenin-related protein in epithelial cells. J. Biol. Chem. 277: 2869-2875. 11711544

Ji, T., Zhang, L., Deng, M., Huang, S., Wang, Y., Pham, T. T., Smith, A. A., Sridhar, V., Cabernard, C., Wang, J. and Yan, Y. (2019). Dynamic MAPK signaling activity underlies a transition from growth arrest to proliferation in Drosophila scribble mutant tumors. Dis Model Mech. 12(8). PubMed ID: 31371383

Katsukawa, M., Ohsawa, S., Zhang, L., Yan, Y. and Igaki, T. (2018). Serpin facilitates tumor-suppressive cell competition by blocking Toll-mediated Yki activation in Drosophila. Curr Biol 28(11):1756-1767. PubMed ID: 29804808

Khoury, M. J. and Bilder, D. (2020). Distinct activities of Scrib module proteins organize epithelial polarity. Proc Natl Acad Sci U S A 117(21): 11531-11540. PubMed ID: 32414916

Ko, J., Fuccillo, M. V., Malenka, R. C. and Sudhof, T. C. (2009). LRRTM2 functions as a neurexin ligand in promoting excitatory synapse formation. Neuron 64(6): 791-798. PubMed ID: 20064387

Kong, D., Zhao, S., Xu, W., Dong, J. and Ma, X. (2022). Fat body-derived Spz5 remotely facilitates tumor-suppressive cell competition through Toll-6-α-Spectrin axis-mediated Hippo activation. Cell Rep 39(12): 110980. PubMed ID: 35732124

Kulshammer, E., Mundorf, J., Kilinc, M., Frommolt, P., Wagle, P. and Uhlirova, M. (2015). Interplay among Drosophila transcription factors Ets21c, Fos and Ftz-F1 drives JNK-mediated tumor malignancy. Dis Model Mech 8: 1279-1293. PubMed ID: 26398940

Laura, R. P., Witt, A. S., Held, H. A., Gerstner, R., Deshayes, K., Koehler, M. F., Kosik, K. S., Sidhu, S. S. and Lasky, L. A. (2002). The Erbin PDZ domain binds with high affinity and specificity to the carboxyl termini of delta-catenin and ARVCF. J. Biol. Chem. 277: 12906-12914. 11821434

Legouis, R., et al. (2000). LET-413 is a basolateral protein required for the assembly of adherens junctions in Caenorhabditis elegans. Nat. Cell Biol. 2: 415-422. PubMed Citation: 10878806

Legouis, R., et al. (2003). Basolateral targeting by leucine-rich repeat domains in epithelial cells. EMBO Rep. 4(11): 1096-102. 14578922

Lehman, K., et al. (1999). Yeast homologues of tomosyn and lethal giant larvae function in exocytosis and are associated with the plasma membrane SNARE, Sec9. J. Cell Biol. 146: 125-140. PubMed Citation: 10402465

Li, M. and Marhold, J. (2001). Differential expression of two scribble isoforms during Drosophila embryogenesis. Mech. Dev. 108: 185-190. 11578873

Li, T., Tian, Y., Li, Q., Chen, H., Lv, H., Xie, W. and Han, J. (2015). The Neurexin/N-Ethylmaleimide-sensitive Factor (NSF) Interaction Regulates Short Term Synaptic Depression. J Biol Chem 290(29): 17656-17667. PubMed ID: 25953899

Logeay, R., Geminard, C., Lassus, P., Rodriguez-Vazquez, M., Kantar, D., Heron-Milhavet, L., Fischer, B., Bray, S. J., Colinge, J. and Djiane, A. (2022). Mechanisms underlying the cooperation between loss of epithelial polarity and Notch signaling during neoplastic growth in Drosophila. Development. PubMed ID: 35005772

Lu H. and Bilder, D. (2005). Endocytic control of epithelial polarity and proliferation in Drosophila. Nat. Cell Biol. 7: 1232-1239. PubMed Citation: 16258546

Mathew, D., et al. (2002). Recruitment of Scribble to the synaptic scaffolding complex requires GUK-holder, a novel dlg binding protein. Curr. Biol. 12: 531-539. 11937021

Matusek, T., Djiane, A., Jankovics, F., Brunner, D., Mlodzik, M. and Mihaly, J. (2006). The Drosophila formin DAAM regulates the tracheal cuticle pattern through organizing the actin cytoskeleton. Development 133: 957-966. PubMed ID: 16469972

Matusek, T., Gombos, R., Szecsenyi, A., Sanchez-Soriano, N., Czibula, A., Pataki, C., Gedai, A., Prokop, A., Rasko, I. and Mihaly, J. (2008). Formin proteins of the DAAM subfamily play a role during axon growth. J Neurosci 28: 13310-13319. PubMed ID: 19052223

Moberg, K. H., et al. (2005). Mutations in erupted, the Drosophila ortholog of mammalian tumor susceptibility gene 101, elicit non-cell-autonomous overgrowth. Dev. Cell 9: 699-710. PubMed Citation: 16256744

Mohr, J., Dash, B.P., Schnoeder, T. M., Wolleschak, D., Herzog, C., Tubio Santamaria, N., Weinert, S., Godavarthy, S., Zanetti, C., Naumann, M., Hartleben, B., Huber, T. B., Krause, D. S., Kahne, T., Bullinger, L. and Heidel, F. H. (2018). The cell fate determinant Scribble is required for maintenance of hematopoietic stem cell function. Leukemia 32(5):1211-1221. PubMed ID: 29467485

Montcouquiol, M., et al. (2003). Identification of Vangl2 and Scrb1 as planar polarity genes in mammals. Nature 423(6936): 173-7. 12724779

Muhammad, K., Reddy-Alla, S., Driller, J. H., Schreiner, D., Rey, U., Bohme, M. A., Hollmann, C., Ramesh, N., Depner, H., Lutzkendorf, J., Matkovic, T., Gotz, T., Bergeron, D. D., Schmoranzer, J., Goettfert, F., Holt, M., Wahl, M. C., Hell, S. W., Scheiffele, P., Walter, A. M., Loll, B. and Sigrist, S. J. (2015). Presynaptic spinophilin tunes neurexin signalling to control active zone architecture and function. Nat Commun 6: 8362. PubMed ID: 26471740

Murdoch, J. N., et al. (2003). Disruption of scribble (Scrb1) causes severe neural tube defects in the circletail mouse. Hum. Mol. Genet. 12(2): 87-98. 12499390

Nagasaka, K., et al. (2006). Human homolog of Drosophila tumor suppressor Scribble negatively regulates cell-cycle progression from G1 to S phase by localizing at the basolateral membrane in epithelial cells. Cancer Sci. 97(11): 1217-25. Medline abstract: 16965391

Nagase, T., et al. (1995). Prediction of the coding sequences of unidentified human genes. IV. The coding sequences of 40 new genes (KIAA0121-KIAA0160) deduced by analysis of cDNA clones from human cell line KG-1. DNA Res. 2: 167-74. PubMed Citation: 8590280

Nagata, R., Nakamura, M., Sanaki, Y. and Igaki, T. (2019). Cell competition is driven by autophagy. Dev Cell 51(1): 99-112. PubMed ID: 31543447

Nelson, K. S., Khan, Z., Molnar, I., Mihaly, J., Kaschube, M. and Beitel, G. J. (2012). Drosophila Src regulates anisotropic apical surface growth to control epithelial tube size. Nat Cell Biol 14: 518-525. PubMed ID: 22446737

Nola, S., et al. (2008). Scrib regulates PAK activity during the cell migration process. Hum. Mol. Genet. 17: 3552-3565. PubMed Citation: 18716323

Ogawa, M., Kawarazaki, Y., Fujita, Y., Naguro, I. and Ichijo, H. (2020). FGF21 Induced by the ASK1-p38 Pathway Promotes Mechanical Cell Competition by Attracting Cells. Curr Biol. PubMed ID: 33357449

Ohtakara, K.,et al. (2002), Densin-180, a synaptic protein, links to PSD-95 through its direct interaction with MAGUIN-1. Genes Cells 7(11): 1149-60. 12390249

Pagliarini, R. A. and Xu. T. (2003), A genetic screen in Drosophila for metastatic behavior. Science 302: 1227-1231. 14551319

Parnas, D., Haghighi, A.P., Fetter, R.D., Kim, S.W. and Goodman, C.S. (2001). Regulation of postsynaptic structure and protein localization by the Rho-type guanine nucleotide exchange factor dPix. Neuron 32: 415-424. 11709153

Petit, M. M., et al. (2005). The tumor suppressor Scrib interacts with the zyxin-related protein LPP, which shuttles between cell adhesion sites and the nucleus. BMC Cell Biol. 6(1): 1. 15649318

Piguel, N. H., et al. (2014). Scribble1/AP2 complex coordinates NMDA receptor endocytic recycling. Cell Rep 9(2):712-27. PubMed ID: 25310985

Poernbacher, I. and Vincent, J. P. (2018). Epithelial cells release adenosine to promote local TNF production in response to polarity disruption. Nat Commun 9(1): 4675. PubMed ID: 30405122

Qin, Y., Capaldo, C., Gumbiner, B. M. and Macara, I. G. (2005). The mammalian Scribble polarity protein regulates epithelial cell adhesion and migration through E-cadherin. J. Cell Biol. 171(6): 1061-71. 16344308

Rhee, D. Y., Cho, D. Y., Zhai, B., Slattery, M., Ma, L., Mintseris, J., Wong, C. Y., White, K. P., Celniker, S. E., Przytycka, T. M., Gygi, S. P., Obar, R. A. and Artavanis-Tsakonas, S. (2014). Transcription factor networks in Drosophila melanogaster. Cell Rep 8: 2031-2043. PubMed ID: 25242320

Roche, J. P., et al. (2002). Regulation of synaptic plasticity and synaptic vesicle dynamics by the PDZ protein Scribble. J. Neurosci. 22(15): 6471-6479. 12151526

Rui, M., Qian, J., Liu, L., Cai, Y., Lv, H., Han, J., Jia, Z. and Xie, W. (2017). The neuronal protein Neurexin directly interacts with the Scribble-Pix complex to stimulate F-actin assembly for synaptic vesicle clustering. J Biol Chem 292(35):14334-14348. PubMed ID: 28710284

Sanaki, Y., Nagata, R., Kizawa, D., Leopold, P. and Igaki, T. (2020). Hyperinsulinemia drives epithelial tumorigenesis by abrogating cell competition. Dev Cell 53(4): 379-389 PubMed ID: 32386602

Stickel, S. and Su, T. T. (2014). Oncogenic mutations produce similar phenotypes in Drosophila tissues of diverse origins. Biol Open 3(3): 201-9. PubMed ID: 24570398

Sun, M., Liu, L., Zeng, X., Xu, M., Liu, L., Fang, M. and Xie, W. (2009). Genetic interaction between Neurexin and CAKI/CMG is important for synaptic function in Drosophila neuromuscular junction. Neurosci Res 64(4): 362-371. PubMed ID: 19379781

Takizawa, S., et al. (2006). Human scribble, a novel tumor suppressor identified as a target of high-risk HPV E6 for ubiquitin-mediated degradation, interacts with adenomatous polyposis coli. Genes Cells 11(4): 453-64. Medline abstract: 16611247

TerBush, T. R., et al. (1996). The Exocyst is a multiprotein complex required for exocytosis in Saccharomyces cerevisiae. EMBO J. 15: 6483-94. PubMed Citation: 8978675

Thompson, B. J. et al. (2005). Tumor suppressor properties of the ESCRT-II complex component Vps25 in Drosophila. Dev. Cell 9: 711-720. PubMed Citation: 16256745

Vaccari, T. and Bilder, D. (2005). The Drosophila tumor suppressor vps25 prevents nonautonomous overproliferation by regulating notch trafficking, Dev. Cell 9: 687-698. PubMed Citation: 16256743

Vandenberg, A. L. and Sassoon, D. A. (2009). Non-canonical Wnt signaling regulates cell polarity in female reproductive tract development via van gogh-like 2. Development 136(9): 1559-70. PubMed Citation: 19363157

Ventura, G., Moreira, S., Barros-Carvalho, A., Osswald, M. and Morais-de-Sa, E. (2020). Lgl cortical dynamics are independent of binding to the Scrib-Dlg complex but require Dlg-dependent restriction of aPKC. Development 147(15). PubMed ID: 32665243

Wada, H., et al. (2005). Dual roles of zygotic and maternal Scribble1 in neural migration and convergent extension movements in zebrafish embryos. Development 132: 2273-2285. 15829519

Walsh, G. S., Grant, P. K., Morgan, J. A. and Moens, C. B. (2011). Planar polarity pathway and Nance-Horan syndrome-like 1b have essential cell-autonomous functions in neuronal migration. Development 138(14): 3033-42. PubMed Citation: 21693519

Wansleeben, C., et al. (2010). Planar cell polarity defects and defective Vangl2 trafficking in mutants for the COPII gene Sec24b. Development 137(7): 1067-73. PubMed Citation: 20215345

Wu, M., Pastor-Pareja, J. C. and Xu, T. (2010). Interaction between RasV12 and scribbled clones induces tumour growth and invasion. Nature 463(7280): 545-8. PubMed Citation: 20072127

Yamben, I. F., Rachel, R. A., Shatadal, S., Copeland, N. G., Jenkins, N. A., Warming, S. and Griep, A. E. (2013). Scrib is required for epithelial cell identity and prevents epithelial to mesenchymal transition in the mouse. Dev Biol 384: 41-52. PubMed ID: 24095903

Yang, C.C., Graves, H.K., Moya, I.M., Tao, C., Hamaratoglu, F., Gladden, A.B. and Halder, G. (2015). Differential regulation of the Hippo pathway by adherens junctions and apical-basal cell polarity modules. Proc Natl Acad Sci USA 112(6):1785-90. PubMed ID: 25624491

Zeitler, J., Hsu, C. P., Dionne, H. and Bilder, D. (2004). Domains controlling cell polarity and proliferation in the Drosophila tumor suppressor Scribble. J. Cell Biol. 167(6): 1137-46. 15611336

Zelhof, A. C., et al. (2001). Drosophila Amphiphysin is implicated in protein localization and membrane morphogenesis but not in synaptic vesicle endocytosis. Development 128: 5005-5015. 11748137

Zhang, H., Webb, D. J., Asmussen, H., Niu, S., and Horwitz, A. F. (2005) A GIT1/PIX/Rac/PAK signaling module regulates spine morphogenesis and synapse formation through MLC. J. Neurosci. 25, 3379-3388. PubMed ID: 15800193

Zhao, M., Szafranski, P., Hall, C. A. and Goode, S. (2008). Basolateral junctions utilize warts signaling to control epithelial-mesenchymal transition and proliferation crucial for migration and invasion of Drosophila ovarian epithelial cells. Genetics 178(4): 1947-71. PubMed Citation: 18430928

Zheng, W., Umitsu, M., Jagan, I., Tran, C. W., Ishiyama, N., BeGora, M., Araki, K., Ohashi, P. S., Ikura, M. and Muthuswamy, S. K. (2016). An interaction between Scribble and the NADPH oxidase complex controls M1 macrophage polarization and function. Nat Cell Biol 18(11): 1244-1252. PubMed ID: 27694890

Žigman, M., et al. (2011). Zebrafish neural tube morphogenesis requires Scribble-dependent oriented cell divisions. Curr. Biol. 21(1): 79-86. PubMed Citation: 21185191


Biological Overview

date revised: 25 August 2023

Home page: The Interactive Fly © 2011 Thomas Brody, Ph.D.