Cbl


REGULATION

An intronic microRNA links Rb/E2F and EGFR signaling

The importance of microRNAs in the regulation of various aspects of biology and disease is well recognized. However, what remains largely unappreciated is that a significant number of miRNAs are embedded within and are often co-expressed with protein-coding host genes. Such a configuration raises the possibility of a functional interaction between a miRNA and the gene it resides in. This is exemplified by the Drosophila melanogaster dE2f1 gene that harbors two miRNAs, mir-11 and mir-998, within its last intron. miR-11 was demonstrated to limit the proapoptotic function of dE2F1 by repressing cell death genes that are directly regulated by dE2F1, however the biological role of miR-998 was unknown. This study shows that one of the functions of miR-998 is to suppress dE2F1-dependent cell death specifically in rbf mutants by elevating EGFR signaling. Mechanistically, miR-998 operates by repressing dCbl, a negative regulator of EGFR signaling. Significantly, dCbl is a critical target of miR-998 since dCbl phenocopies the effects of miR-998 on dE2f1-dependent apoptosis in rbf mutants. Importantly, this regulation is conserved, as the miR-998 seed family member miR-29 repressed c-Cbl, and enhanced MAPK activity and wound healing in mammalian cells. Therefore, the two intronic miRNAs embedded in the dE2f1 gene limit the apoptotic function of dE2f1, but operate in different contexts and act through distinct mechanisms. These results also illustrate that examining an intronic miRNA in the context of its host's function can be valuable in elucidating the biological function of the miRNA, and provide new information about the regulation of the host gene itself (Truscott, 2014. PubMed ID: 25058496).

Protein Interactions

Drosophila Cbl, when expressed in human cultured cells becomes associated with the EGF receptor. Expression of human c-Cbl is accompanied by an EGF-dependent increase in a tyrosine phosphorylated EGF-receptor bound to c-Cbl immunoprecipitates. Transfection with Drosophila Cbl cDNA reveals a p60 polypeptide corresponding to Drosophila Cbl. In nonstimulated cells, Drosophila Cbl is tyrosine phosphorylated, and activation with EGF leads to increased phosphorylation and association of Drosophila Cbl with the EGF-receptor. A Glycine315-to-Glu point mutation mutation in C. elegans Cbl suppresses the ability of this protein to act as a negative regulator of the C. elegans EGF-receptor signaling. The association of Drosophila Cbl and the EGF-receptor is prevented by an analogous mutation in Drosophila Cbl. The analgous mutation totally prevents tyrosine phosphorylation of the expressed protein by the EGF receptor (Meisner, 1997).

The c-Cbl proto-oncogene encodes a multidomain phosphoprotein that has been demonstrated to interact with a wide range of signaling proteins. The biochemical function of c-Cbl in these complexes is, however, unclear. Recent studies with the C. elegans Cbl homolog, sli-1, have suggested that Cbl proteins may act as negative regulators of Egf receptor signaling. Because the Egfr and other protein tyrosine kinase receptor signaling pathways are highly conserved between insects and vertebrates, a Drosophila homolog of c-Cbl was sought for a detailed genetic analysis. Drosophila has a single gene, Cbl, that is homologous to c-cbl. Drosophila Cbl encodes a 52 kDa protein that has a high degree of similarity to c-Cbl and SLI-1 across novel phosphotyrosine-binding (PTB) and RING finger domains. Surprisingly, however, Drosophila Cbl is C-terminally truncated relative to c-Cbl and SLI-1, and consequently is unable to bind SH3-domain containing adaptor proteins, including the Drosophila Grb2 homolog, Drk. Although the Drosophila Cbl protein lacks Drk binding sites it can nevertheless either associate either with a tyrosine phosphorylated protein, or is itself tyrosine phosphorylated in an Egfr dependent manner and it associates with activated Drosophila Egrf receptors in vivo. Consistent with a role for Drosophila Cbl in Egfr dependent patterning in the embryo and adult, Cbl is expressed at a high level in early embryos and throughout the imaginal discs in third instar larvae. This study forms the basis for future genetic analysis of Cbl, aimed at gaining insights into the role of Cbl proteins in signal transduction (Hime, 1997).

Sprouty (Spry) was first identified in a genetic screen in Drosophila as an antagonist of fibroblast and epidermal growth factor receptors and Sevenless signaling, seemingly by inhibiting the receptor tyrosine kinase (RTK)/Ras/MAPK pathway. To date, four mammalian Sprouty genes have been identified; the primary sequences of the gene products share a well conserved cysteine-rich C-terminal domain with their Drosophila counterpart. The N-terminal regions do not, however, exhibit a large degree of homology. This study was aimed at identifying proteins with which human SPRY2 (hSPRY2) interacts in an attempt to understand the mechanism by which Sprouty proteins exert their down-regulatory effects. hSPRY2 associates directly with c-Cbl, a known down-regulator of RTK signaling. A short sequence in the N terminus of hSPRY2 was found to bind directly to the Ring finger domain of c-Cbl. Parallel binding was apparent between the Drosophila homologs of Sprouty and Cbl, with cross-species associations occurring at least in vitro. Coexpression of hSPRY2 abrogates an increase in the rate of epidermal growth factor receptor internalization induced by c-Cbl, whereas a mutant hSPRY2 protein unable to bind c-Cbl showed no such effect. These results suggest that one function of hSPRY2 in signaling processes downstream of RTKs may be to modulate c-Cbl physiological function such as that seen with receptor-mediated endocytosis (Wong, 2001).

In examining the sequence of Drosophila Sprouty, a region was found between amino acids 179 and 199 that shows a low homology to residues 36-53 in hSPRY2. Residues 36-53 lie within region 11-53, which is important for c-Cbl binding. It was therefore investigated if the binding domain could be further refined to a smaller region. FLAG-tagged hSPRY2 truncation and deletion constructs were expressed in 293T cells, and cell lysates were immunoprecipitated with anti-FLAG antibody and probed with anti-c-Cbl antibody. No binding is seen between c-Cbl and the 53C (without residues 1-53) and DeltaN36 (lacking amino acids 36-53) mutants, whereas binding is apparent between c-Cbl and the 30C (without residues 1-30) and full-length constructs. A reciprocal precipitation experiment was performed in which cell lysates from the same transfections were subjected to immunoprecipitation with anti-c-Cbl antibody and immunoblotted with anti-FLAG antibody. The result is in agreement with the above data. Thus, the c-Cbl-binding region of hSPRY2 is contained within sequence 36-53 (Wong, 2001).

To extend the binding investigation to Drosophila Spry and Drosophila Cbl, various constructs were made. (1) To investigate whether the Ring finger domain of Cbl is involved in binding to Spry, 293T cells were transiently transfected with Cbl, dCblDeltaRF (Drosophila Cbl lacking the ring finger domain), or vector alone. Cell lysates were subjected to pull-down assays with GST-Spry or GST alone. Whereas full-length Cbl binds to Spry, dCblDeltaRF does not. This result is indicative of the Ring finger domain of Cbl being involved in its binding to Spry. (2) Binding domain analyses were performed to ascertain the site in Spry that binds to the Ring finger domain of Cbl. The possible involvement of residues 179-199 in Spry was directly addressed. FLAG-tagged full-length Spry and the dSPRYN210 (amino acids 1-210), dSPRY202C (amino acids 202-592), and dSPRYDeltaN179 (mutant with a deletion of amino acids 179-199) constructs were transiently expressed in 293T cells, and lysates were incubated with either GST alone or GST-Cbl. Spry-derived proteins that contain residues 179-199 bind to Cbl, whereas those that lack the sequence do not. The region comprising residues 179-199 of Spry is therefore responsible for its interaction with Cbl; deletion of this region of Spry can similarly abolish its binding to c-Cbl (Wong, 2001).

Cbl and Sprint regulate early steps of RTK endocytosis

Guidance receptors detect extracellular cues and instruct migrating cells how to orient in space. Border cells perform a directional invasive migration during Drosophila oogenesis and use two receptor tyrosine kinases (RTKs), EGFR and PVR (PDGF/VEGF Receptor), to read guidance cues. Spatial localization of RTK signaling within these migrating cells is actively controlled. Border cells lacking Cbl, an RTK-associated E3 ubiquitin ligase, have delocalized guidance signaling, resulting in severe migration defects. Absence of Sprint, a receptor-recruited, Ras-activated Rab5 guanine exchange factor, gives related defects. In contrast, increasing the level of RTK signaling by receptor overexpression or removing Hrs, an endosome-associated, ubiquitin binding protein required for multivesicular body formation and degradation of RTKs, and thereby decreasing RTK degradation, does not perturb migration. Cbl and Sprint both regulate early steps of RTK endocytosis. Thus, a physiological role of RTK endocytosis is to ensure localized intracellular response to guidance cues by stimulating spatial restriction of signaling (Jekely, 2005).

It was reasoned that regulation of RTK turnover might be important to maintain a directional response in border cells, and the effects of mutations likely to affect this process were analyzed. Cbl is a ubiquitin ligase with a conserved role in regulating RTK signaling. Clones of border cells mutant for Cbl are correctly specified, as judged by staining for Slbo, a marker specific for differentiated border cells, but have severe migration defects. To explore the relationship between Cbl and RTK signaling, RTK signaling was manipulated in Cbl mutant border cells. The migration defect in Cbl mutant cells was suppressed by reducing the level of an EGFR ligand (grk/+) and was enhanced by overexpression of either receptor in border cells (UAS-PVR or UAS-EGFR. Note that overexpression of either PVR or EGFR alone has no effect. This indicates that the Cbl phenotype is not due to lack of guidance signaling but instead due to excessive, misregulated RTK signaling (Jekely, 2005).

Consistent with the interpretation that Cbl is required to restrict signaling, the complete failure of many Cbl mutant border cell clusters to migrate resembles the effect of increased guidance receptor signaling due to expression of constitutively active receptors or a strong ligand. In contrast, border cells lacking Pvr and Egfr all eventually initiate migration but never make it to the oocyte. Similar phenotypic effects of manipulating guidance cues have been observed by live imaging of germ cell migration in the zebrafish embryo, a migration guided by a G protein-coupled receptor: migratory cells lacking guidance cues migrate, but randomly, whereas the same cells subject to high uniform guidance cues did not migrate. Thus, border cells lacking Cbl responded as if they were receiving high uniform signaling, a situation that mimics the endpoint of migration (Jekely, 2005).

Mammalian Cbl proteins negatively regulate multiple RTKs by stimulating their ubiquitination and lysosomal degradation. The N-terminal phospho-tyrosine binding domain of Cbl directly binds to activated receptors, and ubiquitin-conjugating enzymes are recruited via the E3 type RING finger. The N-terminal part of Drosophila Cbl also physically interacts with autophosphorylated Pvr. In some assays, these conserved domains are sufficient for mammalian Cbl to regulate Egfr. Cbl can also interact with proteins regulating endocytosis as well as other signaling molecules through its less well conserved C-terminal region. Drosophila Cbl is expressed as two isoforms. Ubiquitous expression of either Cbl-L or Cbl-S, which lacks the C-terminal tail, rescues lethality associated with the Cbl null mutation as well as the migration phenotype. To determine whether E3 ligase activity of Cbl is required, a single cysteine residue essential for this activity was mutated to alanine (Cys-369, corresponds to Cys-381 in human Cbl). The Cbl ring finger mutant was unable to rescue viability of the Cbl mutant or migration of Cbl mutant border cell clones, showing that this function is essential for Cbl activity during migration (Jekely, 2005).

Overall level of RTK activity, in the cell or at the cell cortex, does not need to be precisely controlled to allow migration. Yet Cbl is apparently required to restrict RTK activity. This prompted examination of whether Cbl might affect subcellular localization of RTK signaling at a more refined level. Experiments with Hrs mutants and RTK overexpression suggest that total phospho-tyrosine might be used as a local indicator of RTK signaling. Many proteins are tyrosine-phosphorylated in cells by a number of kinases, but the RTKs have a quantitatively significant effect (direct and indirect). Remarkably, wild-type border cells initiating migration show a clear localization of phospho-tyrosine signal to the front. The front is the side facing the direction of subsequent migration and the source of the ligands, namely the oocyte. Since border cells migrate as a tight cluster of cells, several cells contribute to the front. To further test whether the signal reflected stimulation of endogenous RTKs, a strong Egfr ligand (secreted Spitz) was expressed in border cells to stimulate the endogenous receptor uniformly. This resulted in delocalized phospho-tyrosine signal all over the cortex of the border cells, and, as expected, a block in directed migration (70% nonmigrating clusters). This validated the use of phospho-tyrosine as a reasonable local readout of endogenous RTK activation (Jekely, 2005).

Endogenous Pvr and Egfr are detected at low uniform levels in border cells. Overexpression of Egfr or Pvr results in high level of receptor throughout the cluster; however, the phospho-tyrosine signal remains localized. Thus, local activation is maintained despite RTK overexpression. Consistent with signal location being the critical parameter for guidance signaling, directed migration also proceeds normally upon RTK overexpression. In contrast, border cells mutant for Cbl show a high frequency of delocalized phospho-tyrosine signal. These results indicate that Cbl is important in migrating cells because it is required to restrict RTK signaling spatially within the cell; without Cbl, signaling becomes delocalized. Since Cbl affects RTK endocytosis, whether perturbing endocytosis more generally would have the same effect was tested. Expression of a dominant-negative form of Shibire (dynamin) in border cells initiating migration also caused efficient delocalization of the phospho-tyrosine signal (Jekely, 2005).

The incomplete penetrance of the Cbl phenotype suggests that other molecules might partially compensate for the loss of Cbl. Indirect evidence is available that another potential RTK binding endocytosis regulator called Sprint might have a role in border cells. The mammalian counterpart of Sprint, called RIN1, displays Ras-activated Rab5 guanine nucleotide exchange factor (GEF) activity and can bind Egfr and stimulate Egfr activity. RIN1 also binds and activates the Abelson tyrosine kinase. To analyze the function of Drosophila sprint in vivo, sprint mutants, including a complete loss-of-function mutant, were generated. Despite sprint being the only rin1-related gene in Drosophila, homozygous sprint mutant flies are completely viable and fertile with normal oogenesis. To determine whether Sprint might contribute to regulating RTKs during border cell migration, the cells were challenged by overexpressing Pvr or Egfr in the mutant background. By itself, this overexpression has no effect on migration. In the sprint mutant background, however, RTK overexpression results in significant migration defects and, as for Cbl mutants, a corresponding increase in delocalized phospho-tyrosine signal. This suggests that Sprint might play a role similar to Cbl. Sprint might not be essential under normal conditions due to overlap in function with Cbl. To test this further, border cells mutant for both sprint and Cbl were analyzed. These cells have very severe migration defects and rarely reach the oocyte. For comparison, almost half the Cbl single mutant clusters reach the oocyte. Since sprint has barely any defect on its own, this strong enhancement of the Cbl phenotype is significant. Such a synergistic effect of two null mutants indicates that the gene products function in parallel to regulate the same process (Jekely, 2005).

To understand more about how Sprint might function in vivo, an antibody was generated that detects endogenous Sprint. In a pattern strikingly similar to the wild-type polarized phospho-tyrosine signal, endogenous Sprint was detected at the front of border cells initiating migration. This is consistent with Sprint being recruited to active RTKs. This was confirmed by the ability of overexpressed Egfr or Pvr to recruit endogenous Sprint. In overexpression experiments, it was also found that Sprint has the characteristics expected from its homology to RIN1: Sprint binds Ras-GTP recruited Abelson kinase to the cell cortex and associates with endocytic vesicles. Finally, endogenous Sprint accumulates at the apical cortex of follicle cells, contacting the oocyte, upon transient block of endocytosis. Since endogenous Egfr and Pvr ligands come from the oocyte, this supports the idea that Sprint dynamically associates with early endocytosis of RTKs at the cell cortex. Taken together with the genetic analysis, it is concluded that Cbl and Sprint both serve to maintain RTK signaling localized for guidance, although they stimulate early endocytosis events in molecularly unrelated ways (Jekely, 2005).

Regulators such as Cbl and Sprint might be recruited directly to activated and autophosphorylated RTKs or might bind indirectly, via phosphorylated adaptor proteins. A yeast two-hybrid assay was set up to detect possible direct, phosphorylation-dependent binding. The intracellular domain of Pvr is able to autophosphorylate in yeast and bind SH2 and PTB domain proteins. Binding was detected of both Cbl and Sprint to Pvr, but not a kinase-dead Pvr mutant. Potential docking tyrosines in Pvr were systematically mutated. Mutation of 16 or 14 (YF14) tyrosines results in strong decrease in binding of Cbl and Sprint. To map the binding sites, 5 tyrosines at a time were 'added back' to the YF14 mutant. Although there was no overlap in tyrosines, two of the resulting constructs regained full binding to Cbl and Sprint, indicating that both proteins have more than one direct binding site on Pvr. One construct (YFc) did not bind either Cbl or Sprint directly and therefore seemed to be a potentially useful tool to study the role of their direct binding to Pvr in vivo (Jekely, 2005).

To determine the signaling potential of each Pvr mutant in vivo, the mutations were placed in the context of a constitutively active form of Pvr (λ-Pvr) to induce full, unregulated activation. The ability to block border cell migration and induce F-actin accumulation was monitored. The activity of YF14 was strongly reduced, but each add back mutant had only slightly reduced activity relative to wild-type despite missing nine potential docking tyrosines. Thus, each of the add back mutants was still capable of signaling to affect migration and guidance when artificially activated (Jekely, 2005).

To determine whether the YFc mutations affected receptor regulation, they were placed in the context of full-length Pvr. From transgenes, Pvr and Pvr-YFc were expressed in the ovary at similar levels. As expected, the ability to activate signaling in border cells (measured by anti-dpERK staining) was quantitatively reduced in Pvr-YFc compared to Pvr. This result was confirmed using the sensitive MAP kinase-activated reporter gene kekkon-lacZ. However, expression of Pvr-YFc but not Pvr caused border cell migration defects. Although the frequency of defects was low, finding a gain-of-function activity at all was significant, given that the signaling strength of Pvr-YFc was reduced compared to Pvr. The migration defects were qualitatively similar to those of Cbl mutants and distinct from dominant-negative effects, which even in their strongest form cause migration delays but not arrest. Uniform expression of the ligand PVF1 did not further affect the phenotype of Pvr-YFc, indicating that this form of Pvr that cannot bind Cbl and Sprint had already lost its spatial information. Consistent with this, expression of Pvr-YFc also induced a delocalized phospho-tyrosine signal at a frequency corresponding to the migration defects. This analysis of Pvr itself further indicates that the phenotypes of Cbl and sprint mutant border cells are due to their effects on RTK signaling: recruitment of Cbl and Sprint to Pvr serves to regulate Pvr guidance signaling, specifically to keep it localized (Jekely, 2005).

Thus, Cbl and Sprint are required to keep RTK signaling properly localized. To show this, phospho-tyrosine was used as a read-out of local RTK signaling. Although this reagent is not uniquely specific for the active receptor, the visualized effects of Pvr or Egfr overexpression, Hrs mutation, ligand misexpression, as well as Sprint colocalization, validates its utility in visualizing the high level of local receptor activity found at the leading edge of migrating border cells. The requirement for Cbl and Sprint suggests that the cellular activity required for signal restriction is receptor endocytosis, which is supported by experiments with dominant-negative dynamin. Thus, in this physiological context of guidance by RTKs, receptor endocytosis serves not to downregulate active receptors, but to ensure their correct spatial localization (Jekely, 2005).

The proposed role of RTK endocytosis regulators should be seen in the context of what is already know about RTK signaling and regulation. Signaling from RTKs is initiated upon transphosphorylation of activating tyrosines and docking tyrosines, the latter generating binding sites for PTB and SH2 domain proteins. Receptor activation is elicited by binding of activating ligand but can also occur if two receptor molecules contact each other productively for other reasons. The likelihood of ligand-independent activation depends on receptor density, and hence overexpressed receptors may have ligand-independent activity in addition to responding more strongly to ligands. Inactivation of receptors is therefore critical for proper signaling in the cell. Phosphatases inactivate receptors by catalyzing the reverse reaction of the activation. Phosphatases are very abundant in cells and may be constitutively active. Local inactivation of phosphatases is one mechanism that can lead to spreading of an initially localized RTK signal. In addition, signaling can be inactivated by endocytosis, which leads to degradation of activated receptors, stimulated by molecules such as Cbl and Sprint/RIN1 and at a later step by Hrs (Jekely, 2005).

Most studies of induced endocytosis, in order to give maximal experimental resolution, have been done in tissue culture cells with acute stimulation by high levels of ligand. In tissues, which have steady and modest levels of ligand and a complex, multicellular environment, the role of endocytosis in RTK regulation is less well understood. For example, Egfr signaling is mildly increased in Cbl mutant follicle cell clones, but so mild that even in Cbl, sprint double mutant follicle cells, there are no detectable changes in levels of Egfr, Pvr, or phospho-tyrosine. However, the effects on border cell migration are striking. Receptor proteins do turn over in the tissue and at least some of this turnover is blocked in Hrs mutant cells. However, under physiological conditions in the ovary, the Hrs-dependent degradation is not dependent on ligand and is not required for guided migration. These results show that the physiological role of Cbl and Sprint in border cell guidance is not to control receptor degradation and/or to turn off signaling, but instead to keep the signal localized (Jekely, 2005).

It is becoming appreciated that endocytosis of signaling receptors is not simply a matter of signal attenuation and receptor removal. It was found that RTK endocytosis differentially affected signaling through different pathways, suggesting (1) that signaling can happen in different compartments and (2) that the process of endocytosis could be used to differentially regulate signaling outcomes. For TGF-β signaling, the process of endocytosis actively brings receptors to internal signaling. This study suggests a third role for early aspects of receptor endocytosis in signaling: to keep active signaling complexes localized in the plane of the membrane. This activity prevents signaling from becoming uniform and therefore uninformative about the spatial distribution of the ligand (Jekely, 2005).

How do Cbl and Sprint spatially restrict signaling? They may prevent signaling from becoming delocalized by restricting lateral movement of activated receptors or lateral spread of RTK activation. Microdomains of active RTKs on the plasma membrane or in endocytic pits could maintain activity, whereas they would be inactivated at other places by ubiquitous phosphatases. Alternatively, recycling of activated receptors to new regions of the cell membrane could delocalize signaling. Normally, this recycling might be prevented by Cbl and Sprint activity by routing active RTKs to degradation via the proper endosome compartment (without requiring Hrs). For these two scenarios, however, it is not obvious why physically blocking endocytosis (Shibire dominant-negative) should also delocalize signaling. Shibire/dynamin is a general effector of endocytosis (required for cell viability), and interfering with it therefore is a more blunt tool than manipulating Cbl and Sprint or mutating Pvr. But the effects are unambiguous. This leads to a third hypothesis, whereby endocytosis of active RTKs allows their redelivery or recycling to regions of higher signaling. Endocytosis and plasma membrane redelivery of active proteins contributes to polarization in yeast, another case of controlling spatial information. Obviously, further analysis will be needed to fully explore these cellular mechanisms in vivo. In any case, at sufficiently high level of receptor expression and activation, the regulatory mechanism may collapse. Indeed, when a C-terminally tagged Egfr was expressed at extremely high levels in border cells, migration and phospho-tyrosine staining were perturbed in a manner similar to what was observed for Cbl mutant clones. Like many regulatory mechanisms, the spatial restriction imposed by Cbl and Sprint works effectively within a certain range of input, emphasizing the need for understanding the mechanism of regulation at a physiological range in vivo (Jekely, 2005).

The role of early endocytosis regulators in spatial signal regulation described in this study is clearly physiologically relevant. But how general might it be? The regulation is likely to be relevant when RTKs are used for spatially resolved signaling. RTKs can act as canonical guidance receptors to detect specific ligands. In addition, mammalian Cbl is required for integrin-dependent migration of macrophages and osteoclasts in vitro and in vivo. This requirement was suggested to reflect an active signaling role of Cbl downstream of integrins, but it could also reflect a role for Cbl in localizing signaling analogous to what was seen in border cells. There is ample evidence for cross-talk between integrins and RTKs, which in turn are regulated by Cbl. That RTKs can be activated in the absence of cognate ligand by high receptor density or by cross-talk from other pathways such as integrins might seem at odds with their serving as guidance receptors. But with effective regulatory mechanisms to maintain localized signaling, this excitable signaling system may help migrating cells obtain sufficient sensitivity to read guidance cues over a large dynamic range (Jekely, 2005).

A key issue in guidance signaling is that migrating cells must achieve a polarized output despite having to respond, over a large dynamic range, to subtle concentration differences of an attractant or repellant from one end of the cell to the other. One way to achieve this is to amplify the initial signal difference between stimulation of receptors at the front and back of the cell. The use of PI3 kinase and PTEN phosphatase, two antagonistic enzymes that are reciprocally regulated in Dictyostelium chemotaxis, may be an example of this. Alternatively, guidance cues may simply bias a separate, preexisting intrinsic polarity in the migrating cells. Finally, guidance signaling and intrinsic polarity may interact dynamically to reinforce one another during directed migration. The net outcome is a robust difference between the front and the back of the cell, allowing migration. It is interesting to consider that RTK endocytosis may also function to enhance the difference between signaling in the front and back of migratory border cells. Although the gradient of RTK ligands around border cells can not be measured, it is very unlikely to be as steep as the observed difference in phospho-tyrosine staining. Enhancement of a signaling differential can occur at different levels. Binding of Cbl and Sprint to activated RTKs and recruitment to membrane subdomains may effectively concentrate activated receptors. Due to the density dependence and positive feedback in RTK activation, local activity will then be increased, whereas global inactivation by phosphatases could ensure that signaling is reduced elsewhere. It is suggested that spatial organization of signaling may be controlled by endocytosis and redelivery of active receptors. Such active turnover processes can be used as an effective mechanism to increase signaling differentials within a cell, leading even to spontaneous, or self-organized, polarity. A distinct cell front and cell rear and hence short-term productive migration is often seen in migrating cells even without perception of localized guidance cues, indicating intrinsic polarity. Most migrating cells may need to integrate intrinsic polarity and external guidance. Using a regulatory principle that can produce both intrinsic polarity and local response to guidance cues would provide an elegant means to achieve this (Jekely, 2005).

Differential effects of Cbl isoforms on Egfr signaling in Drosophila

The Cbl family of proteins downregulate epidermal growth factor receptor (Egfr) signaling via receptor internalization and destruction. These proteins contain two functional domains, a RING finger domain with E3 ligase activity, and a proline rich domain mediating the formation of protein complexes. The Drosophila cbl gene encodes two isoforms, D-CblS and D-CblL. While both contain a RING finger domain, the proline rich domain is absent from D-CblS. Expression of either isoform is sufficient to rescue both the lethality of a D-cbl null mutant and the adult phenotypes characteristic of Egfr hyperactivation, suggesting that both isoforms downregulate Egfr signaling. Interestingly, targeted overexpression of D-CblL, but not D-CblS, results in phenotypes characteristic of reduced Egfr signaling and suppresses the effect of constitutive Egfr activation. The level of D-CblL was significantly correlated with the phenotypic severity of reduced Egfr signaling, suggesting that D-CblL controls the efficiency of downregulation of Egfr signaling. Furthermore, reduced dynamin function suppresses the effects of D-CblL overexpression in follicle cells, suggesting that D-CblL promotes internalization of activated receptors. D-CblL is detected in a punctate cytoplasmic pattern, whereas D-CblS is mainly localized at the follicle cell cortex. Therefore, D-CblS and D-CblL may downregulate Egfr through distinct mechanisms (Pai, 2006).

D-CblL has domains that allow it to act both as an ubiquitin ligase and as an adaptor mediating protein–protein interaction through its proline rich regions, while D-CblS lacks the proline rich region in the C terminus that may promote receptor internalization. Both isoforms rescue lethality in a D-cbl mutant background and can control Egfr signaling roughly correctly in the wings, eyes, and follicle cells. A low level of D-CblS partially rescues lethality and is accompanied by a mild wing phenotype characteristic of hyperactivated Egfr. However, when the expression levels are sufficient for better survival, no patterning defects are observed. This full rescue suggests interactions mediated through the C-terminus of D-CblL are not essential for viability, and D-CblS can downregulate activated Egfr properly by itself. Indeed, a mutant encoding a similar protein in C. elegans, sli-1ΔC, also retains some ability to negatively regulate Egfr signaling during vulval development of C. elegans. Furthermore, Cbl-3, which does not bind to CIN85, promotes the ubiquitination and degradation of activated Egfr in mammals (Pai, 2006).

Although the levels of D-CblS are critical for animal and cell survival as indicated by the lethal effect produced by high ubiquitous expression of transgenes driven by e22cGal4 or heat shock, no or only a slight patterning defect is observed when D-CblS is overexpressed using tissue specific Gal4 lines in the wings, eyes and egg shells. These data have demonstrated that D-CblS has the ability to control Egfr signaling in a manner similar to the wild-type D-cbl gene and high expression levels of D-CblS reduce survival but exert very little or no effects on patterning in specific tissues. Therefore, D-CblS appears to have a higher dynamic range than D-CblL in controlling proper Egfr signaling, which may reflect its higher level of expression in vivo compared to D-CblL (Pai, 2006).

Internalization and degradation of receptors is an efficient way to terminate signaling, and protein complexes interacting with the C-terminus of Cbl are involved in ligand-induced downregulation of Egfr through internalization. For example, the C terminal proline rich region of Cbl interacts with the multiple SH3 domain-containing protein, Grb2, which is required for Egfr endocytosis. Grb2 has been demonstrated to regulate the rate of Egfr internalization through clathrin-coated pits by recruiting c-Cbl. The Drosophila homolog of Grb2, Drk, binds to several kinds of proline rich motif through its SH3 domains. Interestingly, one classic SH3 binding motif PXXPXR was found in the C terminus of D-CblL. Moreover, two such motifs exist in Sli-1, which showed genetic interactions with SEM5, the C. elegans homolog of Grb2. Therefore, it is possible that D-CblL more efficiently promotes Egfr degradation in part through association with Drk, which does not bind to D-CblS (Pai, 2006).

In an alternative mechanism leading to Egfr downregulation, the Cbl-CIN85-endophilin complex mediates ligand-induced internalization of Egfr as well. The CIN85-Endophilin complex is thought to play a critical role at invagination during endocytosis by inducing negative membrane curvature. The PXXXPR motif in Cbl, responsible for interaction with CIN85, is also present in D-CblL. Through an intensive data base search a putative Drosophila homolog of CIN85 (CG31012), was identified which has three SH3 domains in the N-terminus and a proline rich region prior to the coiled-coil domain at the C-terminus. This information suggests that a protein–protein interaction network for Cbl-CIN85-Endophilin exists in Drosophila (Pai, 2006).

The phenotype characteristic of Egfr hypoactivation generated in the eyes, wings and egg shells by overexpression of D-CblL demonstrates that D-CblL is critical for control of Egfr signaling and high levels of D-CblL elevate the downregulation of Egfr signaling. Indeed it was also found that the severity of the ventralized egg shell phenotype is dependent on the levels of D-CblL. In addition, a dominant negative shibire mutant suppressed the D-Cbl effects on egg shell. These data indicate that activated Egfr units can be internalized rapidly through the activity of the endocytic protein complexes when D-CblL, but not D-CblS, is present at high levels. In addition to patterning defects, overexpression of D-CblL is usually accompanied by a lethal effect. The rapid internalization may lead to hypoactivation of Egfr and thus result in lethality, since Egfr signaling is required for cell survival (Pai, 2006).

It is crucial to control both the strength and the duration of signaling during development, and internalization of receptors is a key process for this control. To date, many reports have shown that there are several redundant mechanisms of internalization of activated receptors, such as clathrin-dependent and -independent, dynamin-dependent and -independent processes, as well as caveolar endocytosis. The contribution of these pathways in Egfr downregulation in vivo is not completely understood (Pai, 2006).

The data have shown that D-CblS, which lacks the C terminal proline rich domain, regulates Egfr signaling properly, suggesting that the interactions between Cbl and adaptor proteins are dispensable. D-CblS presumably promotes internalization of activated Egfr solely through its E3 ligase activity, and ubiquitination of Egfr subsequently recruits ubiquitin binding proteins, such as Epsin family members, which then serve as clathrin or caveolin adaptors to form clathrin-coated vesicle or caveolae in endocytic pathways. In contrast, D-CblL may have additional endocytic pathways other than those that are ubiquitination dependent, such as the interaction with the CIN85-endophilin complex through its C terminus. In addition, the Drk binding site in the C terminus of D-CblL may also increase the association of D-CblL and activated Egfr. Therefore, increase of D-CblL levels may promote Egfr downregulation by the lysosomal pathway mediated through D-CblL-CIN85-endophilin complex. In addition, D-CblL may promote polyubiquitination of Egfr through its E3 ligase activity while the constitutive binding between its SH3 domain and the proline rich motif of Drk provides a stable association between activated receptor and D-CblL, thus polyubiquitinated Egfr can be degraded by the 26S proteasome. In contrast, D-CblS only allows monoubiquitination on activated Egfr, which allows lysosomal degradation (Pai, 2006).

It is known that post-translational modifications of both receptor and components of the endocytic machinery control the rate of receptor degradation. The EGFR complex is sorted into multivesicular bodies where it either fuses with the lysosome for complete degradation and termination of signaling, or is recycled which prolongs the signaling. Since, the punctate pattern of D-CblL staining indicates a possible subcellular vesicle localization, D-CblL may also promote sorting activated Egfr to the lysosome by ubiquitination modification on components of the endocytic machinery. Taken together, these data strongly suggest that CblS and CblL mediate internalization of activated Egfr through distinct mechanisms, which are characterized by different genetic behaviors. Given the different intracellular localization patterns of D-CblS and D-CblL it is very likely that these differences reflect distinct endocytic pathways, which regulate Egfr signaling with different efficiencies and in a partially redundant manner (Pai, 2006).


DEVELOPMENTAL BIOLOGY

Members of the Cbl family of proteins act as E3 ubiquitin-protein ligases and have been associated with the down regulation of a variety of receptor tyrosine kinsases. Cbl proteins associate with many different cell signaling molecules, suggesting that they may have functions outside of the RING finger-mediated ubiquitin ligase activity. The Drosophila Cbl gene encodes two splice forms. These isoforms are expressed differently during Drosophila embryogenesis. Both isoforms are maternally expressed but the long isoform of Cbl is also transiently expressed in the invaginating mesoderm and later is specifically expressed in neurons of the central nervous system (CNS). Cbl protein is shown to be localized to axons of the longitudinal connectives and commissures in the central nervous system (Hime, 2001).

To begin to determine if the two Drosophila Cbl proteins have distinct roles in development the expression patterns of the isoforms were examined. Since the two mRNA isoforms have different 3' untranslated regions, isoform specific antisense riboprobes could be generated. Rabbit antisera to two regions of Cbl were prepared. Antiserum GH2 was generated against a glutathione S-transferase::CblS fusion protein and GH6 against a region of the CblL specific exon fused to GST. GH2 immunoprecipitates both CblS and CblL from in vitro translated proteins and GH6 specifically precipitated CblL. CblL expression in vivo could not be detected using GH6, so subsequent immunolocalization was conducted using GH2. Western blots of lysates from S2 cells expressing enhanced yellow fluorescent protein (YFP) alone and a CblS::YFP fusion protein show that GH2 can specifically detect the fusion protein (Hime, 2001).

CblS is strongly maternally expressed with high levels of mRNA in preblastoderm embryos. A concentration of cblS mRNA was observed at the posterior pole of several preblastoderm embryos, although high levels were not observed in pole cells. CblS mRNA remains at high levels up until cellular blastoderm when it begins to be degraded. Subsequently, only a low level of expression is observed throughout the later stages of embryogenesis, for example. The level of expression is significantly higher than background levels detected with a sense-strand riboprobe and may reflect low level zygotic expression (Hime, 2001).

CblL has a dynamic expression pattern during early embryogenesis and neurogenesis. A high level of maternal CblL expression is present analogous to CblS. In contrast to CblS, CblL mRNA is completely absent by cellular blastoderm stage. No association with the posterior pole is evident. CblL is first zygotically expressed during gastrulation in invaginating mesoderm. However, this expression is transient and rapidly downregulated postgastrulation. A low level of expression is present in germband extended embryos. CblL is next observed in stage 11 embryos in the CNS. Expression is specific to developing neurons with no evidence of expression in more ventral layers of the nerve cord (i.e., neuroblasts). Levels of CblL mRNA remain relatively constant but more cells of the ventral nerve cord are observed to express CblL as the nervous system develops. In stage 15/16 embryos CblS is widely expressed in the ventral nerve cord and brain CblS.

Cbl protein is localized to axons of the central nervous system. The GH2 antisera was used to examine localization of Cbl protein within the CNS. In stage 13 embryos a low level of staining is evident across all cells of the embryo. This probably reflects the low level zygotic expression of cblS observed from mRNA in situ hybridization. A higher level of expression is present in the longitudinal connectives of the CNS. Expression is also observed in axons of the brain. The mRNA in situ data would suggest that axonal localization of CblL is detected. Axon staining is more intense in stage 16 embryos. Dissection of the CNS from a stage 16 embryo shows that Cbl protein is localized to the commissures as well as the longitudinal connectives, suggesting that most CNS axons express the protein (Hime, 2001).

Effects of Mutation or Deletion

Myopic acts in the endocytic pathway to enhance signaling by the Drosophila EGF receptor

Endocytosis of activated receptors can control signaling levels by exposing the receptors to novel downstream molecules or by instigating their degradation. Epidermal growth factor receptor (EGFR) signaling has crucial roles in development and is misregulated in many cancers. Myopic, the Drosophila homolog of the Bro1-domain tyrosine phosphatase HD-PTP, promotes EGFR signaling in vivo and in cultured cells. myopic is not required in the presence of activated Ras or in the absence of the ubiquitin ligase Cbl, indicating that it acts on internalized EGFR, and its overexpression enhances the activity of an activated form of EGFR. Myopic is localized to intracellular vesicles adjacent to Rab5-containing early endosomes, and its absence results in the enlargement of endosomal compartments. Loss of Myopic prevents cleavage of the EGFR cytoplasmic domain, a process controlled by the endocytic regulators Cbl and Sprouty. It is suggested that Myopic promotes EGFR signaling by mediating its progression through the endocytic pathway (Miura, 2008).

The Epidermal growth factor receptor (EGFR) is required for cell differentiation and proliferation in numerous developmental systems, and activation of the human EGFR homologs, ERBB1-4, is implicated in many cancers. EGFR signaling events are terminated following removal of the receptor from the cell membrane by endocytosis. Ubiquitylation of EGFR by Cbl, an E3 ubiquitin ligase, initiates its internalization into clathrin-coated vesicles and its transit through early and late endosomes, which differ by the exchange of Rab7 for Rab5. EGFR can either return to the cell surface in Rab11-containing recycling endosomes, or reach the lysosome for degradation. Delivery of receptors to the lysosome requires sorting from the limiting membrane of late endosomes into the internal vesicles of multivesicular bodies (MVBs), a process mediated by four endosomal sorting complexes required for transport (ESCRT-0, I, II and III). Ubiquitylated receptors are bound by Hepatocyte growth factor regulated tyrosine kinase substrate (Hrs) in ESCRT-0, Tumor susceptibility gene 101 (Tsg101; also known as Erupted) in ESCRT-I and Vacuolar protein sorting 36 (Vps36) in ESCRT-II, and their deubiquitylation and internalization are coordinated by ESCRT-III (Miura, 2008).

Genetic or pharmacological blocks of endocytosis prevent degradation of EGFR and other receptors. In Drosophila, Hrs mutations block MVB invagination, trapping receptor tyrosine kinases (RTKs) and other receptors on the outer membrane of the MVB, and sometimes leading to enhanced signaling. Mutations in the ESCRT complex subunits Tsg101 (ESCRT-I) and Vps25 (ESCRT-II) cause overproliferation owing to the accumulation of mitogenic receptors such as Notch and Thickveins. In mammalian cells, loss of Hrs (also known as Hgs) or Tsg101 results in increased EGFR signaling. However, other studies have demonstrated a positive role for endocytosis in receptor signaling. Mutations affecting the Drosophila trafficking protein Lethal giant discs dramatically increase Notch signaling only in the presence of Hrs, indicating that signaling is maximized at a specific point in the endocytic process. Wingless (Wg) signaling is enhanced by internalization into endosomes, where it colocalizes with downstream signaling molecules. In mammalian cells, EGFR encounters the scaffolding proteins Mek1 partner (Mp1) and p14, which are required for maximal phosphorylation of the downstream component mitogen-activated protein kinase (MAPK), only on endosomes (Miura, 2008).

The characterization of the novel Drosophila gene myopic (mop) is described here. Loss of mop affects EGFR-dependent processes in eye and embryonic development, and reduces MAPK phosphorylation by activated EGFR in cultured cells. Mop acts upstream of Ras activation to promote the function of activated, internalized EGFR. Mop is homologous to human HD-PTP (PTPN23 - Human Gene Nomenclature Database) (Toyooka, 2000), which contains a Bro1 domain that is able to bind the ESCRT-III complex component SNF7 (CHMP4B - Human Gene Nomenclature Database) (Ichioka, 2007; Kim, 2005) and a tyrosine phosphatase domain. Mop is present on intracellular vesicles, and cells lacking mop have enlarged endosomes and reduced cleavage of the EGFR cytoplasmic domain. It is proposed that Mop potentiates EGFR signaling by enhancing its progression through endocytosis. Consistent with this hypothesis, it was found that components of the ESCRT-0 and ESCRT-I complexes are also required for EGFR signaling in Drosophila cells (Miura, 2008).

This study shows that Mop is necessary for EGFR signaling in vivo and in cultured cells. Mop is located on endosomes and affects endosome size, promotes cleavage of EGFR and lysosomal entry of its ligand, and is not required in the absence of the Cbl or Sty proteins that regulate endocytic trafficking of EGFR. These data suggest that Mop enhances EGFR signaling by facilitating its progression through the endocytic pathway. Consistent with this model, Hrs and ESCRT-I subunits also have a positive effect on EGFR signaling (Miura, 2008).

The Bro1 domain of yeast Bro1 is sufficient for localization to late endosomes through its binding to the ESCRT-III subunit Snf7 (Kim, 2005), and this domain is present in many proteins involved in endocytosis. Bro1 itself is required for transmembrane proteins to reach the vacuole for degradation; it promotes protein deubiquitylation by recruiting and activating Doa4, a ubiquitin thiolesterase. Since mutations in the E3 ubiquitin ligase gene Cbl can rescue mop mutant clones, recruiting deubiquitylating enzymes might be one of the functions of Mop. The vertebrate Bro1-domain protein Alix (also known as AIP1 and Pdcd6ip) inhibits EGFR endocytosis by blocking the ubiquitylation of EGFR by Cbl, and by preventing the binding of Ruk (Sh3kbp1), which recruits endophilins, to the EGFR-Cbl complex. However, CG12876, not Mop, is the Drosophila ortholog of Alix (Miura, 2008).

A closer vertebrate homolog of Mop, which has both Bro1 and tyrosine phosphatase domains, has been named HD-PTP in human (Toyooka, 2000) and PTP-TD14 in rat. HD-PTP shares with Alix the ability to bind Snf7 and Tsg101, but does not bind to Ruk (Ichioka, 2007). PTP-TD14 was found to suppress cell transformation by Ha-Ras, and required phosphatase activity for this function (Cao, 1998). The activity of Mop described in this study appears distinct in that Mop acts upstream of Ras activation, and no requirement was demonstrated for the catalytic cysteine in its predicted phosphatase domain. If Mop does act as a phosphatase, Hrs would be a candidate substrate because tyrosine phosphorylation of Hrs by internalized receptors promotes its degradation, and Hrs levels appear reduced in mop mutant clones (Miura, 2008).

Endocytosis has been proposed to play several different roles in receptor signaling. Most commonly, endocytosis followed by receptor degradation terminates signaling. However, endocytosis can also prolong the duration of signaling or influence its subcellular location. Receptors may also signal through different downstream pathways localized to specialized endosomal compartments (Miura, 2008).

Genetic studies in Drosophila have emphasized the importance of endocytic trafficking for receptor silencing. Mutations in Hrs, Vps25 or Tsg101 result in the accumulation of multiple receptors on the perimeter membrane of the MVB, leading to enhanced signaling. Depletion of Hrs or Tsg101 in mammalian cells also results in increased EGFR signaling, although the two molecules have distinct effects on MVB morphology. By contrast, this study found that mop and Hrs mutants exhibit diminished EGFR signaling in vivo, and depletion of mop, Tsg101 or Vps28 reduces EGFR signaling in S2 cells. Progression through the endocytic pathway may thus be required for maximal EGFR signaling, at least in some contexts (Miura, 2008).

Several possible mechanisms could explain such a requirement for endocytic progression. MAPK phosphorylation may be enhanced in the presence of signaling components present on late endosomes. Cleavage of the EGFR cytoplasmic domain, which requires Mop activity, might enhance EGFR signaling. The cleaved intracellular domain of ErbB4 has been shown to enter the nucleus and regulate gene expression (Sardi, 2006), suggesting the possibility that Mop affects a nuclear function of EGFR in addition to promoting MAPK phosphorylation. Alternatively, the reduction in EGFR signaling in mop mutants could be due to a failure to recycle the receptor to the cell surface. Mutations in the yeast Vps class C genes, which are required for trafficking to late endosomes, also prevent the recycling of cargo proteins. Recycling is essential for EGFR-induced proliferation of mammalian cells, and may promote the localized RTK signaling that drives directional cell migration (Miura, 2008).

Despite the reduction in EGFR signaling in mop mutants, signaling by other receptors such as Notch, Smoothened and Torso is unaffected. This phenotypic specificity could be due to a dedicated function of Mop in the EGFR pathway, or to high sensitivity of EGFR signaling to a general process that requires Mop. Although the Mop-related protein Alix has been found in a complex with EGFR (Schmidt, 2004), no physical interaction of Mop with EGFR could be detected. The function of mop is not limited to promoting EGFR signaling; it also promotes trafficking of Wg and expression of the Wg target gene sens. In addition, mop is required for normal cellularization of the embryo, and its cellularization phenotype is not rescued by removal of Cbl (data not shown) (Miura, 2008). Additional studies will be required to determine whether all endosomes, or only a specific subclass, are affected by mop. Interestingly, EGF treatment of mammalian cells induces EGFR trafficking through a specialized class of MVBs. Although significant colocalization of activated EGFR with Mop was not seen, EGFR may transiently pass through Mop-containing endosomes before accumulating in another compartment. The wing disc appears less sensitive than the eye disc to the effect of mop on EGFR signaling. This might be due to differences in the endogenous levels of Cbl or other mediators of EGFR internalization, or in the strength or duration of signaling necessary to activate target genes, or to the use of a different ligand with distinct effects on receptor trafficking (Miura, 2008).

Taken together, these results identify a positive role for progress through the endocytic pathway and for the novel molecule Mop in EGFR signaling in Drosophila. The importance of upregulation of the trafficking proteins Rab11a, Rab5a and Tsg101 for EGFR signaling in hepatomas and breast cancers highlights the potential value of specific effectors of EGFR endocytosis as targets for anti-cancer therapies (Miura, 2008).

Neuronal Cbl controls biosynthesis of insulin-like peptides in Drosophila melanogaster

The Cbl family proteins function as both E3 ubiquitin ligases and adaptor proteins to regulate various cellular signaling events, including the insulin/insulin-like growth factor 1 (IGF1) and epidermal growth factor (EGF) pathways. These pathways play essential roles in growth, development, metabolism, and survival. This study shows that in Drosophila Cbl (dCbl) regulates longevity and carbohydrate metabolism through downregulating the production of Drosophila insulin-like peptides (dILPs) in the brain. dCbl is highly expressed in the brain and knockdown of the expression of dCbl specifically in neurons by RNA interference increases sensitivity to oxidative stress or starvation, decreased carbohydrate levels, and shortened life span. Insulin-producing neuron-specific knockdown of dCbl results in similar phenotypes. dCbl deficiency in either the brain or insulin-producing cells upregulates the expression of dilp genes, resulting in elevated activation of the dILP pathway, including phosphorylation of Drosophila Akt and Drosophila extracellular signal-regulated kinase (dERK). Genetic interaction analyses revealed that blocking Drosophila epidermal growth factor receptor (dEGFR)-dERK signaling in pan-neurons or insulin-producing cells by overexpressing a dominant-negative form of dEGFR abolishes the effect of dCbl deficiency on the upregulation of dilp genes. Furthermore, knockdown of c-Cbl in INS-1 cells, a rat β-cell line, also increases insulin biosynthesis and glucose-stimulated secretion in an ERK-dependent manner. Collectively, these results suggest that neuronal dCbl regulates life span, stress responses, and metabolism by suppressing dILP production and the EGFR-ERK pathway mediates the dCbl action. Cbl suppression of insulin biosynthesis is evolutionarily conserved, raising the possibility that Cbl may similarly exert its physiological actions through regulating insulin production in β cells (Yu, 2012).

Akap200 suppresses the effects of Dv-cbl expression in the Drosophila eye

The Drosophila orthologue of the c-Cbl proto-oncogene acts to downregulate signalling from receptor tyrosine kinases by enhancing endocytosis of activated receptors. Expression of an analogue of the C-terminally truncated v-Cbl oncogene, Dv-cbl, in the developing Drosophila eye conversely leads to excess signalling and disruption to the well-ordered adult compound eye. Co-expression of activated Ras with Dv-cbl leads to a severe disruption of eye development. A transposon-based inducible expression system was used to screen for molecules that can suppress the Dv-cbl phenotype and an allele was identified that upregulates the A-kinase anchoring protein, Akap200. Overexpression of Akap200 not only suppresses the phenotype caused by Dv-cbl expression, but also the severe disruption to eye development caused by the combined expression of Dv-cbl and activated Ras. Akap200 is also endogenously expressed in the developing Drosophila eye at a level that modulates the effects of excessive signalling caused by expression of Dv-cbl (Sannang, 2012).


REFERENCES

Andoniou, C. E., Thien, C. B. and Langdon, W. Y. (1994). Tumour induction by activated abl involves tyrosine phosphorylation of the product of the cbl oncogene. EMBO J. 13 (19): 4515-4523. PubMed Citation: 7925293

Andoniou, C. E., Thien, C. B. and Langdon, W. Y. (1996). The two major sites of cbl tyrosine phosphorylation in abl-transformed cells select the crkL SH2 domain. Oncogene 12(9): 1981-9. PubMed Citation: 8649859

Bhat, A., et al. (1997). Interactions of CBL with BCR-ABL and CRKL in BCR-ABL-transformed myeloid cells. J. Biol. Chem. 272(26): 16170-16175. PubMed Citation: 9195915

Buday, L., et al. (1996). Interactions of Cbl with two adapter proteins, Grb2 and Crk, upon T cell activation. J. Biol. Chem. 271 (11): 6159-6163. PubMed Citation: 8626404

Cao, L., Zhang, L., Ruiz-Lozano, P., Yang, Q., Chien, K. R., Graham, R. M. and Zhou, M. (1998). A novel putative protein-tyrosine phosphatase contains a BRO1-like domain and suppresses Ha-ras-mediated transformation. J. Biol. Chem. 273: 21077-21083. PubMed Citation: 9694860

Chiusaroli, R., et al. (2003). Deletion of the gene encoding c-Cbl alters the ability of osteoclasts to migrate, delaying resorption and ossification of cartilage during the development of long bones. Dev. Biol. 261: 537-547. 14499658

Choi, J. H., et al. (2003). Cbl competitively inhibits epidermal growth factor-induced activation of phospholipase C-gamma1. Mol. Cells 15(2): 245-55. 12803489

Crowley, M. R., Bowtell, D. and Serra, R. (2005). TGF-beta, c-Cbl, and PDGFR-alpha the in mammary stroma. Dev. Biol. 279(1): 58-72. 15708558

de Jong, R., et al. (1995). Crkl is complexed with tyrosine-phosphorylated Cbl in Ph-positive leukemia. J. Biol. Chem. 270: 21468-21471. PubMed Citation: 7545163

Donovan, J. A., et al. (1994). The protein product of the c-cbl protooncogene is the 120-kDa tyrosine-phosphorylated protein in Jurkat cells activated via the T cell antigen receptor. J. Biol. Chem. 269 (37): 22921-22924. PubMed Citation: 8083187

Fournel, M., et al. (1996). Association of tyrosine protein kinase Zap-70 with the protooncogene product p120c-cbl in T lymphocytes. J. Exp. Med. 183(1): 301-6. PubMed Citation: 8551236

Fukazawa, T., et al. (1995). The SH3 domain-binding T cell tyrosyl phosphoprotein p120. Demonstration of its identity with the c-cbl protooncogene product and in vivo complexes with Fyn, Grb2, and phosphatidylinositol 3-kinase. J. Biol. Chem. 270: 19141-19150. PubMed Citation: 7642581

Galisteo, M. L., et al. (1995). Tyrosine phosphorylation of the c-cbl proto-oncogene protein product and association with epidermal growth factor (EGF) receptor upon EGF stimulation. J. Biol. Chem. 270: 20242-20245. 7657591

Graham, L. J., et al. (1998). Sequences surrounding the Src-homology 3 domain of phospholipase Cgamma-1 increase the domain's association with Cbl. Biochem. Biophys. Res. Commun. 249(2): 537-41. PubMed Citation: 9712732

Haglund, K., et al. (2005). Sprouty2 acts at the Cbl/CIN85 interface to inhibit epidermal growth factor receptor downregulation. EMBO Rep. 6(7): 635-41. 15962011

Hall, A. B., et al. (2003). hSpry2 is targeted to the ubiquitin-dependent proteasome pathway by c-Cbl. Curr. Biol. 13: 308-314. 12593796

Hanazono, Y., et al. (1996). Proto-oncogene products Vav and c-Cbl are involved in the signal transduction through Grb2/Ash in hematopoietic cells. Acta Haematol. 95: 236-242. PubMed Citation: 8677749

Hartley, D., Meisner, H. and Corvera, S. (1995). Specific association of the beta isoform of the p85 subunit of phosphatidylinositol-3 kinase with the proto-oncogene c-cbl. J. Biol. Chem. 270(31): 18260-3. PubMed Citation: 7629144

Hime, G. R., et al. (1997). D-Cbl, the Drosophila homologue of the c-Cbl proto-oncogene, interacts with the Drosophila EGF receptor in vivo, despite lacking C-terminal adaptor binding sites. Oncogene 14(22): 2709-19. PubMed Citation: 9178769

Hime, G. R., et al. (2001). Dynamic expression of alternate splice forms of D-cbl during embryogenesis. Mech. Dev. 102: 235-238. 11287200

Huang, F. and Sorkin, A. (2005). Growth factor receptor binding protein 2-mediated recruitment of the RING domain of Cbl to the epidermal growth factor receptor is essential and sufficient to support receptor endocytosis. Mol. Biol. Cell 16(3): 1268-81. 15635092

Ichioka, F., Takaya, E., Suzuki, H., Kajigaya, S., Buchman, V. L., Shibata, H. and Maki, M. (2007). HD-PTP and Alix share some membrane-traffic related proteins that interact with their Bro1 domains or proline-rich regions. Arch. Biochem. Biophys. 457: 142-149. PubMed Citation: 17174262

Jekely, G., Sung, H. H., Luque, C. M. and Rorth, P. (2005). Regulators of endocytosis maintain localized receptor tyrosine kinase signaling in guided migration. Dev. Cell 9(2): 197-207. 16054027

Joazeiro, C. A., et al. (1999). The tyrosine kinase negative regulator c-Cbl as a RING-type, E2-dependent ubiquitin-protein ligase. Science 286: 309-312. PubMed Citation: 10514377

Kaabeche, K., et al. (2005). Cbl-mediated ubiquitination of alpha5 integrin subunit mediates fibronectin-dependent osteoblast detachment and apoptosis induced by FGFR2 activation. J. Cell Sci. 118(Pt 6): 1223-32. 15728256

Kim, J., Sitaraman, S., Hierro, A., Beach, B. M., Odorizzi, G. and Hurley, J. H. (2005). Structural basis for endosomal targeting by the Bro1 domain. Dev. Cell 8: 937-947. PubMed Citation: 15935782

Kitanaka, A., et al. (1996). CD38-mediated growth suppression of B-cell progenitors requires activation of phosphatidylinositol 3-kinase and involves its association with the protein product of the c-cbl proto-oncogene. Blood 88: 590-598. PubMed Citation: 8695807

Kontani, K., et al. (1996). Tyrosine phosphorylation of the c-cbl proto-oncogene product mediated by cell surface antigen CD38 in HL-60 cells. J. Biol. Chem. 271: 1534-1537. PubMed Citation: 8576149

Lee, P. S., et al. (1999). The Cbl protooncoprotein stimulates CSF-1 receptor multiubiquitination and endocytosis, and attenuates macrophage proliferation. EMBO J. 18(13): 3616-28. PubMed Citation: 10393178

Levkowitz, G., et al. (1999). Ubiquitin ligase activity and tyrosine phosphorylation underlie suppression of growth factor signaling by c-Cbl/Sli-1. Mol. Cell 4(6): 1029-40. PubMed Citation: 10635327

Lill, N. L., et al. (2000). The evolutionarily conserved N-terminal region of Cbl is sufficient to enhance down-regulation of the epidermal growth factor receptor. J. Biol. Chem. 275(1): 367-77. PubMed Citation: 10617627

Liu, Y. C., et al. (1997). Serine phosphorylation of Cbl induced by phorbol ester enhances its association with 14-3-3 Proteins in T cells via a novel serine-rich 14-3-3-binding Motif. J. Biol. Chem. 272 (15): 9979-9985

Mason, J. M., et al. (2004). Tyrosine phosphorylation of Sprouty proteins regulates their ability to inhibit growth factor signaling: a dual feedback loop. Mol. Biol. Cell. 15(5): 2176-88. 15004239

Meisner, H. and Czech, M. P. (1995a). Coupling of the proto-oncogene product c-Cbl to the epidermal growth factor receptor. J. Biol. Chem. 270: 25332-25335 (1995)

Meisner, H., et al. (1995b). Interactions of Cbl with Grb2 and phosphatidylinositol 3'-kinase in activated Jurkat cells. Mol Cell Biol 15 (7): 3571-3578

Meisner, H., et al. (1997). Interactions of Drosophila Cbl with Epidermal growth factor receptors and role of Cbl in R7 photoreceptor cell development. Mol. Cell. Biol 2217-25

Meng, F. and Lowell, C. A. (1998). A beta1 integrin signaling pathway involving Src-family kinases, Cbl and PI-3 kinase is required for macrophage spreading and migration EMBO J. 17: 4391-4403

Meng, W., et al. (1999). Structure of the amino-terminal domain of Cbl complexed to its binding site on ZAP-70 kinase. Nature 398(6722): 84-90

Mohapatra, B., Zutshi, N., An, W., Goetz, B., Arya, P., Bielecki, T. A., Mustaq, I., Storck, M. D., Meza, J. L., Band, V. and Band, H. (2017). An essential role of CBL and CBL-B ubiquitin ligases in mammary stem cell maintenance. Development [Epub ahead of print]. PubMed ID: 28100467

Miura, G. I., Roignant, J. Y., Wassef, M. and Treisman, J. E. (2008). Myopic acts in the endocytic pathway to enhance signaling by the Drosophila EGF receptor. Development 135(11): 1913-22. PubMed Citation: 18434417

Miyake, S., et al. (1998). The tyrosine kinase regulator Cbl enhances the ubiquitination and degradation of the platelet-derived growth factor receptor alpha. Proc. Natl. Acad. Sci. 95(14): 7927-7932

Ota, S., et al. (2000). The RING finger domain of Cbl is essential for negative regulation of the Syk tyrosine kinase. J. Biol. Chem. 275(1): 414-22

Ota, Y. and Samelson, L. E. (1997). The product of the proto-oncogene c-cbl: A negative regulator of the Syk tyrosine kinase. Science 276: 418-420

Pai, L.-M., Barcelo, G. and Schüpbach, T. (2000). D-cbl, a negative regulator of the Egfr pathway, is required for dorsoventral patterning in Drosophila oogenesis. Cell 103: 51-61.

Pai, L. M., et al. (2006). Differential effects of Cbl isoforms on Egfr signaling in Drosophila. Mech. Dev. 123(6): 450-62. 16844358

Panchamoorthy, G., et al. (1996). p120cbl is a major substrate of tyrosine phosphorylation upon B cell antigen receptor stimulation and interacts in vivo with Fyn and Syk tyrosine kinases, Grb2 and Shc adaptors, and the p85 subunit of phosphatidylinositol 3-kinase. J. Biol. Chem. 271: 3187-3194

Rao, N., et al. (2001). The non-receptor tyrosine kinase Syk is a target of Cbl-mediated ubiquitylation upon B-cell receptor stimulation. EMBO J. 20: 7085-7095. 11742985

Reedquist, K. A., et al. (1996). Stimulation through the T cell receptor induces Cbl association with Crk proteins and the guanine nucleotide exchange protein C3G. J. Biol. Chem. 271: 8435-8442

Ribon, V., et al. (1996). The product of the cbl oncogene forms stable complexes in vivo with endogenous Crk in a tyrosine phosphorylation-dependent manner. Mol. Cell. Biol. 16: 45-52

Ribon, V., et al. (1998). A novel, multifuntional c-Cbl binding protein in insulin receptor signaling in 3T3-L1 adipocytes. Mol. Cell. Biol. 18(2): 872-879

Rubin, C., et al. (2003). Sprouty fine-tunes EGF signaling through interlinked positive and negative feedback loops. Curr. Biol. 13: 297-307. 12593795

Safford, M., et al. (2005). Egr-2 and Egr-3 are negative regulators of T cell activation. Nat. Immunol. 6(5): 472-80. 15834410

Salgia, R., et al. (1996). p210 BCR/ABL induces formation of complexes containing focal adhesion proteins and the protooncogene product p120c-Cbl

Sannang, R. T., et al. (2012). Akap200 suppresses the effects of Dv-cbl expression in the Drosophila eye. Mol. Cell. Biochem. 369(1-2): 135-45. PubMed Citation: 22773306

Sanjay, A., et al. (2001). Cbl associates with Pyk2 and Src to regulate Src kinase activity, alphavß3 integrin-mediated signaling, cell adhesion, and osteoclast motility. J. Cell Bio. 152: 181-196. 11149930

Sardi, S. P., Murtie, J., Koirala, S., Patten, B. A. and Corfas, G. (2006). Presenilin-dependent ErbB4 nuclear signaling regulates the timing of astrogenesis in the developing brain. Cell 127: 185-197. PubMed Citation: 17018285

Sattler, M., et al. (1996). The proto-oncogene product p120CBL and the adaptor proteins CRKL and c-CRK link c-ABL, p190BCR/ABL and p210BCR/ABL to the phosphatidylinositol-3' kinase pathway. Oncogene 12: 839-846

Sattler, M., et al. (1997). Differential signaling after beta1 integrin ligation is mediated through binding of CRKL to p120(CBL) and p110(HEF1). J. Biol. Chem. 272 (22): 14320-14326

Sawasdikosol, S., et al. (1996). Tyrosine-phosphorylated Cbl binds to Crk after T cell activation. J. Immunol. 157: 110-116

Schmidt, M. H., Hoeller, D., Yu, J., Furnari, F. B., Cavenee, W. K., Dikic, I. and Bogler, O. (2004). Alix/AIP1 antagonizes epidermal growth factor receptor downregulation by the Cbl-SETA/CIN85 complex. Mol. Cell. Biol. 24: 8981-8993. PubMed Citation: 15456872

Senechal, K., et al. (1998). Structural requirements for function of the Crkl adapter protein in fibroblasts and hematopoietic cells. Mol. Cell. Biol. 18(9): 5082-90

Shishido, T., et al. (2000). The kinase-deficient Src acts as a suppressor of the Abl kinase for Cbl phosphorylation. Proc. Natl. Acad. Sci. 97: 6439-6444

Smit, L., van der Horst, G. and Borst, J. (1996). Formation of Shc/Grb2- and Crk adaptor complexes containing tyrosine phosphorylated Cbl upon stimulation of the B-cell antigen receptor. Oncogene 13: 381-389

Soubeyran, P., et al. (2002). Cbl-CIN85-endophilin complex mediates ligand-induced downregulation of EGF receptors. Nature 416: 183-187. 11894095

Teckchandani, A. M., et al. (2005). The multidomain protooncogenic protein c-Cbl binds to tubulin and stabilizes microtubules. Exp. Cell Res. 306(1): 114-27. 15878338

Thien, C. B. F., Walker, F. and Langdon, W. Y. (2001). RING finger mutations that abolish c-Cbl-directed polyubiquitination and downregulation of the EGF receptor are insufficient for cell transformation. Molec. Cell 7: 355-365

Toyooka, S., Ouchida, M., Jitsumori, Y., Tsukuda, K., Sakai, A., Nakamura, A., Shimizu, N. and Shimizu, K. (2000). HD-PTP: A novel protein tyrosine phosphatase gene on human chromosome 3p21.3. Biochem. Biophys. Res. Commun. 278: 671-678. PubMed Citation: 11095967

Truscott, M., Islam, A. B., Lightfoot, J., Lopez-Bigas, N. and Frolov, M. V. (2014). An intronic microRNA links Rb/E2F and EGFR signaling. PLoS Genet 10: e1004493. PubMed ID: 25058496

Uemura, N., et al. (1997). The BCR/ABL oncogene alters interaction of the adapter proteins CRKL and CRK with cellular proteins. Leukemia 11 (3): 376-385

Waterman, H., et al. (2002). A mutant EGF-receptor defective in ubiquitylation and endocytosis unveils a role for Grb2 in negative signaling EMBO J. 21: 303-313. 11823423

Wong, A., Lamothe, B., Lee, A., Schlessinger, J. and Lax, I. (2004). FRS2 alpha attenuates FGF receptor signaling by Grb2-mediated recruitment of the ubiquitin ligase Cbl. Proc. Natl. Acad. Sci. 99(10): 6684-9. 11997436

Wong, E. S. M., et al. (2001). Evidence for direct interaction between Sprouty and Cbl. J. Biol. Chem. 276: 5866-5875. 11053437

Wong, E. S. M., et al. (2002). Sprouty2 attenuates epidermal growth factor receptor ubiquitylation and endocytosis, and consequently enhances Ras/ERK signaling. EMBO J. 21: 4796-4808. 12234920

Xiong, H., et al. (2005). Ubiquitin-dependent degradation of interferon regulatory factor-8 mediated by Cbl down-regulates interleukin-12 expression. J. Biol. Chem. 280(25): 23531-9. 15837792

Yokouchi, M., et al. (1999). Ligand-induced ubiquitination of the epidermal growth factor receptor involves the interaction of the c-Cbl RING finger and UbcH7. J. Biol. Chem. 274(44): 31707-12. PubMed Citation: 10531381

Yoon, C. H., et al. (1995). Similarity of sli-1, a regulator of vulval development in C. elegans, to the mammalian proto-oncogene c-cbl. Science 269: 1102-1105. PubMed Citation: 7652556

Yu, Y., et al. (2012). Neuronal Cbl controls biosynthesis of insulin-like peptides in Drosophila melanogaster. Mol. Cell Biol. 32(18): 3610-23. PubMed Citation: 22778134

Zhang, Z., et al. (1999). A direct interaction between the adaptor protein cbl-b and the kinase zap-70 induces a positive signal in T cells. Curr. Biol. 9(4): 203-6. PubMed Citation: 10074432

Zheng, N., et al. (2000). Structure of a c-Cbl-UbcH7 complex: RING domain function in ubiquitin-protein ligases. Cell 102(4): 533-9. 10966114


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

date revised: 25 October 2014 

Home page: The Interactive Fly © 1997 Thomas B. Brody, Ph.D.

The Interactive Fly resides on the
Society for Developmental Biology's Web server.