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

Gene name - saxophone

Synonyms -

Cytological map position - 42C1-F8

Function - surface receptor

Keyword(s) - dpp pathway, dorsal ventral polarity

Symbol - sax

FlyBase ID:FBgn0003317

Genetic map position - 2-[57]

Classification - TGF beta receptor

Cellular location - surface



NCBI links: Entrez Gene

sax orthologs: Biolitmine
Recent literature
Patrnogic, J., Heryanto, C. and Eleftherianos, I. (2018). Transcriptional up-regulation of the TGF-beta intracellular signaling transducer Mad of Drosophila larvae in response to parasitic nematode infection. Innate Immun: 1753425918790663. PubMed ID: 30049242
Summary:
The common fruit fly Drosophila melanogaster is an exceptional model for dissecting innate immunity. However, knowledge on responses to parasitic nematode infections still lags behind. Recent studies have demonstrated that the well-conserved TGF-beta signaling pathway participates in immune processes of the fly, including the anti-nematode response. To elucidate the molecular basis of TGF-beta anti-nematode activity, a transcript level analysis of different TGF-beta signaling components was performed following infection of D. melanogaster larvae with the nematode parasite Heterorhabditis gerrardi. No significant changes were found in the transcript level of most extracellular ligands in both bone morphogenic protein (BMP) and activin branches of the TGF-beta signaling pathway between nematode-infected larvae and uninfected controls. However, extracellular ligand, Scw, and Type I receptor, Sax, in the BMP pathway as well as the Type I receptor, Babo, in the activin pathway were substantially up-regulated following H. gerrardi infection. The results suggest that receptor up-regulation leads to transcriptional up-regulation of the intracellular component Mad in response to H. gerrardi following changes in gene expression of intracellular receptors of both TGF-beta signaling branches. These findings identify the involvement of certain TGF-beta signaling pathway components in the immune signal transduction of D. melanogaster larvae against parasitic nematodes.
Moulton, M. J., Humphreys, G. B., Kim, A. and Letsou, A. (2020). O-GlcNAcylation Dampens Dpp/BMP Signaling to Ensure Proper Drosophila Embryonic Development. Dev Cell 53(3): 330-343. PubMed ID: 32369743
Summary:
BMP (bone morphogenetic protein) signaling activity is precisely controlled by both pathway agonists and antagonists. This study identify a previously unrecognized BMP signaling antagonist. The Drosophila BMP type I receptor Sax (Saxophone) functions as a Dpp (Decapentaplegic) receptor in Drosophila embryos, but that its activity is normally inhibited by the O-linked glycosyltransferase Sxc (Super sex combs). In wild-type embryos, Sax activity is inhibited, and the BMP type I receptor Tkv (Thickveins) is the sole conduit for Dpp. In contrast, in sxc mutants, the Dpp signal is transduced by both Tkv and Sax, and elevated Dpp signaling results in embryonic lethality. This study also demonstrates that Sxc O-glycosylates Sax and observe elevated Dpp signaling in response to maternal restriction of dietary sugar. These findings link fertility to nutritive environment and point to Sax signaling as the nutrient-sensitive branch of BMP signaling.
Kramer, J., Neves, J., Koniikusic, M., Jasper, H. and Lamba, D. A. (2021). Dpp/TGFβ-superfamily play a dual conserved role in mediating the damage response in the retina. PLoS One 16(10): e0258872. PubMed ID: 34699550
Summary:
Retinal homeostasis relies on intricate coordination of cell death and survival in response to stress and damage. Signaling mechanisms that coordinate this process in the adult retina remain poorly understood. This study identified Decapentaplegic (Dpp) signaling in Drosophila and its mammalian homologue Transforming Growth Factor-beta (TGFβ) superfamily, that includes TGFβ and Bone Morphogenetic Protein (BMP) signaling arms, as central mediators of retinal neuronal death and tissue survival following acute damage. Using a Drosophila model for UV-induced retinal damage, this study showed that Dpp released from immune cells promotes tissue loss after UV-induced retinal damage. Interestingly, a dynamic response of retinal cells was found to this signal: in an early phase, Dpp-mediated stimulation of Saxophone/Smox signaling promotes apoptosis, while at a later stage, stimulation of the Thickveins/Mad axis promotes tissue repair and survival. This dual role is conserved in the mammalian retina through the TGFβ/BMP signaling, as supplementation of BMP4 or inhibition of TGFβ using small molecules promotes retinal cell survival, while inhibition of BMP negatively affects cell survival after light-induced photoreceptor damage and NMDA induced inner retinal neuronal damage. These data identify key evolutionarily conserved mechanisms by which retinal homeostasis is maintained.
BIOLOGICAL OVERVIEW

Decapentaplegic is responsible for induction of dorsal-ventral polarity in the fly. Imagine the effects of its removal: the back of the fly fails to develop and becomes instead a neurogenic ectoderm resembling tissue usually found in the ventral portion of the trunk. The resulting fly would have two fronts and no back.

Saxophone, along with Thick veins and Punt, constitute the Drosophila receptor for decapentaplegic. These receptors mediate the transduction of DPP signals into the cell. Mutations in these genes have the same effect as mutation in dpp, since without DPP's receptors, its signals fail to be communicated to the cells that should receive them.

Saxophone has an intracellular kinase domain and a conserved extracellular cysteine reside structure. Kinases add phosphate resides to target proteins. This phosphorylation event is the main process by which receptors pass signals between proteins; the transfer of signals to downstream effectors. Along with Thickveins, SAX is homologous to type I TGF-beta receptors.

sax appears to be expressed more ubiquitously but required less ubiquitously than Thick veins, and SAX requires the function of both Thick veins and Punt (Wharton, 1995, and Ruberte, 1995). Since sax is only necessary for the specification of the dorsalmost cell fate (amnioserosa) which requires the highest levels of DPP activity, SAX might serve to interpret peak levels of DPP. Thus additional type I receptors for DPP, such as SAX, probably function to augment or modify a response to DPP mediated by the primary receptors encoded by Punt and TKV (Ruberte, 1995).

In Drosophila wing discs, a morphogen gradient of Dpp has been proposed to be a determinant of the transcriptional response thresholds of the downstream genes spalt (sal) and optomotor blind (omb). Evidence is presented that the concentration of the type I receptor Tkv must be low to allow long-range Dpp diffusion. However, low Tkv receptor concentrations result in low signaling activity. To enhance signaling at low Dpp concentrations, a second ligand, Tgf-beta-60A, has been found to augment Dpp/Tkv activity. Tgf-beta-60A signals primarily through the type I receptor Sax, which synergistically enhances Tkv signaling and is required for proper Omb expression. Omb expression in wing discs is found to require synergistic signaling by multiple ligands and receptors to overcome the limitations imposed on Dpp morphogen function by receptor concentration levels (Haerry, 1998).

The phenotypic consequences of overexpressing constitutively active forms of Tkv and Sax receptors in the developing wing was investigated using the GAL4-UAS system. The A9-Gal4 line was used: this drives high-level expression of Gal4 in the entire wing disc before it is restricted to the dorsal pouch at late third instar stage. In wild-type discs, the Sal and Omb products are symmetrically expressed along the anterior/posterior (A/P) boundary in response to Dpp. Normally, the Sal domain is restricted to cells in the wing pouch that are in close proximity to the Dpp-expressing cells, while Omb responds to lower levels of Dpp and is expressed in cells further away from the A/P boundary. The anterior boundary of Sal has been shown to specify the location where the longitudinal vein 2 (L2) is formed, while the formation of L5 coincides approximately with the posterior boundary of the Omb domain, but a causal relationship has not yet been established. When Dpp is ubiquitously expressed in wing discs, they become overgrown and the expression of both Sal and Omb is expanded. Like Dpp, overexpression of constitutively active Tkv (TkvA) also leads to disc overgrowth and ectopic induction of Sal and Omb. All cells in wings derived from animals expressing either Dpp or activated Tkv appear to differentiate into vein tissue, as exemplified by production of vein-specific morphological markers such as dark pigment and longer bristles. In contrast to TkvA, expression of either one or two copies of SaxA or development at 30°C, which results in an approximately twofold increase of Gal4 activity, is not sufficient to expand either Sal or Omb and produces only weak adult phenotypes consisting primarily of ectopic and thickened veins with a small amount of wing blistering in the region of the posterior cross vein. This phenotype is similar to that seen in animals raised at 18°C, which express low levels of TkvA. Although these findings suggest that Sax function may be qualitatively similar to that of Tkv but simply weaker, higher levels of activated Sax (four copies) still cannot mimic the effects of activated Tkv, such as the expansion of Sal and Omb (Haerry, 1998).

When high levels of activated Sax activity are combined with low levels of activated Tkv, the result is more than additive. The combination of one copy of saxA and low levels of Tkv leads to overgrowth, with the expansion of Omb (but not Sal), and results in a strong wing phenotype. The interaction of Sax and Tkv is synergistic. Taken together, these data suggest that Sax and Tkv synergistically interact and control the expression of a common target gene, omb. Activation of omb expression requires a level of signaling that can be activated by either high levels of Tkv activity alone or by a synergistic interaction between low levels of Tkv and high levels of Sax activity. In contrast, Sal activation requires a higher level of signaling, which can only be achieved by high levels of Tkv activity (Haerry, 1998).

Since both Tkv and Sax, as well as the type II receptor Put, have been implicated in mediating Dpp signaling, whether the loss in signal activity of these receptors would cause similar patterning defects in the wing was investigated. If these three receptors all bind the same ligand and signal to the same sets of downstream genes, it would be expected that a reduction in the activity of any individual receptor should result in qualitatively similar phenotypes that differ in severity only. Increasing levels of dominant negative receptors were expressed in different regions of the developing wing disc. Similar to using an allelic series of hypomorphic mutations, it was expected that expression of increasing copy numbers of dominant negative receptors should result in progressively more severe phenotypes. Ubiquitous expression of 3-4 copies of either form of two dominant negative Tkv1 constructs results in small wings with partial loss of L4 and both cross veins. In addition, L2 and L3 are closer together and the triple-row margin bristles are shifted more distally/posteriorly, as expected if the level of Dpp signal is reduced by titration of Dpp into nonproductive complexes. At higher levels (6-8 copies) of dominant negative Tkv1, very small adult wings are produced that show fusion of L2 and L3 as well as L4 and L5. Similar phenotypes are produced by expressing dominant negative versions of the alternative Tkv isoform that have an N-terminal extended extracellular domain, and also by expression of dominant negative Put. Both the Sal and the Omb domains are strongly reduced: the adult wings show fusion of L2 with L3 and L4 with L5. The wing phenotypes obtained with increasing levels of dominant negative Tkv and Put resemble those of certain combinations of dpp loss-of-function alleles, which is consistent with the notion that Dpp is primarily signaling through the combination of the Tkv and Put receptors. In contrast to these observations, dominant negative Sax constructs produce different results. When increasing copy numbers (1-8 copies) of dominant negative Sax are expressed, the discs become smaller and the Omb domain is reduced to the size of the normal Sal domain. But unlike expressing dominant negative Tkv, the Sal domain is not affected. In the adult wing, L5 and the posterior cross vein are lost compared to losing L3 and L4 after expression of dominant negative Tkv or Put. In addition, L2 is shifted more proximally and the proximal triple-row bristles that expand more distally/posteriorly in dominant negative Tkv wings are replaced by more proximal costa bristles. While the distance between L3 and L4 is normal, the overall shape of the wing becomes more ‘strap-like’, suggesting loss of peripheral tissue rather than the central tissue that is deleted in animals expressing Tkv or Put dominant negative receptors. These results suggest that dominant negative Sax acts in a qualitatively different manner from dominant negative Tkv (Haerry, 1998).

These results indicate that while the reduction of Tkv and Put activity affects the whole disc (Sal, Omb and growth), the expression of dominant negative Sax only affects the peripheral region of the disc (Omb and peripheral growth). If the dominant negative receptors function primarily by titrating Dpp, then it is curious why the overexpression phenotypes of dominant negative Sax are different. One possibility is that these receptors do not simply signal in response to Dpp but also in response to the binding of other ligands as well. Of the other two BMP-type ligands that have been described in Drosophila, scw shows no detectable expression at this stage. However, Tgf-beta-60A is expressed broadly in wing discs, and mutant analyses indicate that Tgf-beta-60A is required for normal wing development. Given its role in wing patterning, the effects of heteroallelic Tgf-beta-60A mutations were examined on Sal and Omb expression. Similar to discs expressing dominant negative Sax, Sal expression in Tgf-beta-60A mutant discs is normal while the Omb domain is reduced, particularly in the dorsal compartment. These observations are consistent with the notion that a second BMP-type ligand, Tgf-beta-60A, is required in addition to Dpp for proper Omb expression. Furthermore, the similarity of the Tgf-beta-60A loss-of-function and the dominant negative Sax phenotypes is consistent with recently described genetic interactions between Tgf-beta-60A and sax mutations and suggests that Tgf-beta-60A could signal in part through Sax (Haerry, 1998).

Ubiquitous overexpression of moderate levels of Tgf-beta-60A does not result in excessive disc overgrowth and does not alter the distribution of Sal and Omb. The resulting wings are slightly larger and exhibit minor venation defects along L2 and L5. However, similar to Dpp or TkvA, higher levels of Tgf-beta-60A overexpression expands both Sal and Omb and results in blistered and pigmented adult wings. Since only activated Tkv but not Sax is able to expand Sal and Omb expression, these findings are consistent with the notion that expression of moderate levels of Tgf-beta-60A leads to signaling preferentially through Sax, producing relative mild phenotypes, while higher concentrations of Tgf-beta-60A may also result in signaling through Tkv, producing phenotypes similar to activated Tkv (Haerry, 1998).

An investigation was carried out to determine if Tgf-beta-60A contributes to wing development primarily in the form of homodimers or Tgf-beta-60A/Dpp heterodimers. Results: (1) the level of Tgf-beta-60A mRNA appears to be significantly less than that of DPP, based on RNA in situ hybridization, indicating that heterodimers are not likely to be very abundant assuming similar translational efficiencies. (2) Localized overexpression of Tgf-beta-60A in the dpp-expressing cells does not result in any mutant phenotypes. (3) Expression of Tgf-beta-60A in the posterior compartment results in overgrowth, an expansion of the Sal and Omb domains, and restriction all adult wing defects exclusively to the posterior compartment. Since Tgf-beta-60A expression in this experiment does not overlap with Dpp-secreting cells, no Dpp/Tgf-beta-60A heterodimers should form, since heterodimer formation requires expression of both proteins in the same cell. Therefore, Tgf-beta-60A functions most likely as a homodimer. This finding is consistent with recent genetic analysis showing that clones of Tgf-beta-60A mutant cells that do not include dpp-expressing cells nevertheless produce patterning defects. It has been shown that dominant negative Tkv is more potent than Sax for inhibiting Dpp signaling, while dominant negative Sax is a stronger suppressor than Tkv of Tgf-beta-60A signaling. High levels of Tkv receptor limit Dpp diffusion and restrict Omb expression (Haerry, 1998).

A novel, noncanonical BMP pathway modulates synapse maturation at the Drosophila neuromuscular junction

At the Drosophila NMJ, BMP signaling is critical for synapse growth and homeostasis. Signaling by the BMP7 homolog, Gbb, in motor neurons triggers a canonical pathway-which modulates transcription of BMP target genes, and a noncanonical pathway-which connects local BMP/BMP receptor complexes with the cytoskeleton. This study describes a novel noncanonical BMP pathway characterized by the accumulation of the pathway effector, the phosphorylated Smad (pMad), at synaptic sites. Using genetic epistasis, histology, super resolution microscopy, and electrophysiology approaches, it was demonstrated that this novel pathway is genetically distinguishable from all other known BMP signaling cascades. This novel pathway does not require Gbb, but depends on presynaptic BMP receptors and specific postsynaptic glutamate receptor subtypes, the type-A receptors. Synaptic pMad is coordinated to BMP's role in the transcriptional control of target genes by shared pathway components, but it has no role in the regulation of NMJ growth. Instead, selective disruption of presynaptic pMad accumulation reduces the postsynaptic levels of type-A receptors, revealing a positive feedback loop which appears to function to stabilize active type-A receptors at synaptic sites. Thus, BMP pathway may monitor synapse activity then function to adjust synapse growth and maturation during development (Sulkowski, 2016).

BMPs fulfill multiple functions during NMJ development via canonical and noncanonical pathways. In motor neurons, signaling by Gbb triggers a canonical BMP signaling that regulates transcription of BMP target genes and a noncanonical BMP pathway that connects Wit with LIMK1 and the cytoskeleton. This study describes a novel non-canonical BMP pathway, which induces selective accumulation of pMad at presynaptic sites. This pathway does not require Gbb, but depends on presynaptic BMP receptors Wit and Sax and postsynaptic GluRIIA. This novel pathway does not contribute to the NMJ growth and instead appears to set up a positive feedback loop that modulates the postsynaptic distribution of type-A and type-B receptors as a function of synapse activity (Sulkowski, 2016).

At the Drosophila NMJ, BMP signaling controls NMJ growth and promotes synapse homeostasis. BMP fulfills all these functions via canonical and noncanonical pathways. Canonical BMP signaling activates presynaptic transcriptional programs with distinct roles in the structural and functional development of the NMJ. For example, the BMP pathway effector Trio can rescue NMJ growth in BMP pathway mutants, but does not influence synapse physiology, whereas Target of Wit (Twit) can partially restore the mini frequency but has no effect on NMJ growth. It has been shown that both muscle and neuron derived Gbb are required for the structural and functional integrity of NMJ, and multiple mechanisms that regulate Gbb expression, secretion and extracellular availability have been described. Binding of Gbb to its receptors also triggers a noncanonical, Mad-independent pathway that requires the C-terminal domain of Wit. This domain is conserved among Drosophila Wit and vertebrate BMPRII and functions to recruit and activate cytoskeletal regulators such as LIMK1. In flies, Wit-mediated activation of LIMK1 mediates synapse stability and enables rapid, activity-dependent synaptic growth (Eaton, 2005; Piccioli, 2014; Sulkowski, 2016 and references therein).

This study uncovered a novel, noncanonical BMP pathway that triggers accumulation of presynaptic pMad in response to postsynaptic GluRIIA receptors. This pathway requires Wit and Sax, suggesting that various BMP pathways compete for shared components. Super resolution imaging mapped the pMad domains at active zones, in close proximity to the presynaptic membrane. These domains concentrate the pMad immunoreactivities into thin discs that reside mostly within individual synapse boundaries. The size and shape of pMad domains suggest that pMad could associate with membrane-anchored complexes at the active zone. Since BMP signals are generally short lived, these pMad domains likely represent pMad that, upon phosphorylation, remains associated with the BMP/BMPR kinase complexes at synaptic sites. Alternatively, pMad may accumulate in synaptic aggregates that protect it from dephosphorylation. While the second possibility cannot be excluded, several lines of evidence support the first scenario. First, excess Mad cannot increase the levels of synaptic pMad. Second, neuronal expression of activated Tkv/Sax but not Mad can restore the synaptic pMad at Importin impβ11 mutant NMJs. Finally, during neural tube closure, junctional pSmad1/5/8 and its association with PAR complexes depend on BMPs. Previous studies indicate a reduction of synaptic pMad signals in response to muscle-specific Mad RNAi. This study too has observed such a reduction. In addition, this study found a significant decrease of postsynaptic IIA/IIB ratio in Mad-depleted muscles: GluRIIA and GluRIIB synaptic levels were reduced to 49% and respectively 78% of control. Since GluRIIA is key to the synaptic pMad accumulation it is suspected that the muscle Mad RNAi phenotype is due to perturbation in synaptic GluRIIA levels, perhaps by interference with the Activin signaling pathway (Sulkowski, 2016).

How are the BMP/BMPR complexes stabilized at synaptic sites? Studies on single receptors demonstrate that the confined mobility of BMPRI on the plasma membrane is key to stabilize BMP/BMPR complexes and differentially stimulate canonical versus noncanonical signaling. Direct interactions between phosphorylated Smad5 and the Par3-Par6-aPKC polarity complex occur at the apical junctions. Similarly, synaptic pMad, which remains associated with BMP/BMPR complexes, may engage in interactions that restrict the mobility of BMP/BMPR complexes on the presynaptic membrane. Nemo-mediated phosphorylation of Mad-S25 could disrupt the pMad/BMPR association and expose the BMP/BMPR complexes, so they could dissociate and/or be internalized. The heteromeric BMPR complexes are transient; ligand binding greatly increases their lifespan and stability. Albeit Gbb is not essential for synaptic pMad, it may act redundantly with other ligands to stabilize BMP/BMPR local complexes. Several ligands secreted in the synaptic cleft have been shown to bind and signal via BMPRII; they include glia secreted Maverik, Myoglianin, which could be secreted from muscle and/or glia, and Activins. However, these ligands also appear to signal via a canonical Activin pathway, which regulates the postsynaptic GluRIIA/GluRIIB abundance at the Drosophila NMJ. Alterations in the Activin signaling pathway drastically alter the synaptic recruitment of both iGluR subtypes, in particular the GluRIIA, which controls synaptic pMad, making it difficult to identify the nature and the directionality of the signaling molecule(s) involved in the synaptic pMad accumulation. Interestingly, all of these ligands are substrates for BMP-1/Tolloid-type enzymes, which control their activity and distribution. Treatments that induce long-term stimulation up-regulate a BMP-1/Tolloid homolog in Aplysia neurons (Sulkowski, 2016).

An intriguing aspect of this novel BMP pathway is the dependence on active postsynaptic GluRIIA, which is both required and sufficient for pMad accumulation at active zones. Since pMad and BMP/BMPR complexes cluster at synaptic sites, it is speculated that trans-synaptic complexes may couple postsynaptic type-A glutamate receptors with presynaptic BMP/BMPRs. The synaptic cleft is 200 Å; the iGluR tetramer expands 135 Å in the synaptic cleft, and the BMP/BMPR complexes ~55 Å. The iGluRs auxiliary subunit Neto has extracellular CUB and LDLa domains predicted to expand 120-130 Å in the synaptic cleft, based on related structures. CUB domains are BMP binding motifs that may localize BMP activities and/or facilitate ligand binding to BMPRs. In this model, Neto provides the link between postsynaptic GluRIIA and presynaptic BMP/BMPR complexes. During receptors gating cycle, the iGluRs undergo corkscrew motions that shorten the channels as revealed by cryo-electron microscopy. Such movements may influence the stability of trans-synaptic complexes and allow synaptic pMad to function as a sensor for GluRIIA activity (Sulkowski, 2016).

While more components of this novel pathway remain to be determined, it is clear that this pathway does not contribute to NMJ growth and instead has a critical role in synapse maturation. Unlike canonical BMP signaling, loss of local pMad induces minor reductions in bouton number and does not rescue the NMJ overgrowth of endocytosis mutants. Local pMad accumulates independently of Wit-mediated LIMK1 activation and does not appear to influence synapse stabilization; in fact, nrx mutants have synapse adhesion defects but show increased synaptic pMad levels. The striking correlation between synaptic pMad levels and GluRIIA activity, together with previous findings that GluRIIA activity and gating behavior directly impacts receptor mobility and synaptic stabilization suggest a positive feedback mechanism in which active GluRIIA receptors induce stabilization of BMP/BMPR complexes at synaptic sites which, in turn, promote stabilization of type-A receptors at PSDs. In this scenario, presynaptic pMad marks active BMP/BMPR complexes and acts to maintain the local BMP/BMPR complexes in large clusters that evade endocytosis. Selective disruption of local pMad via a neuronal dominant-negative MadS25D presumably destabilizes the large presynaptic BMP/BMPR clusters and causes a significant reduction in the IIA/IIB ratio and quantal size (Sulkowski, 2016).

This positive feedback couples synaptic activity with synapse development and is controlled by (1) active GluRIIA receptors, (2) presynaptic BMP receptors, Wit, Sax, and likely Tkv, (3) mechanisms regulating BMPR heteromers assembly, endocytosis and turnover, and (4) the ability of pMad to remain associated with its own kinase upon phosphorylation. Perturbations of any of these components trigger variations in local pMad levels accompanied by changes in the IIA/IIB ratio and/or quantal size. For example, nemo mutants have increased synaptic pMad levels and increased mEJCs, while imp mutants have decreased synaptic pMad levels and decreased mEJPs. The assembly and function of these putative trans-synaptic complexes, in particular ligand availability, should be influenced by the composition and organization of the synaptic cleft. Indeed, local pMad and quantal size are increased in mutants lacking heparan sulfate 6-O-endosulfatase (sulf1), or 6-O-sulfotransferase (hs6st). Since this Mad-dependent, noncanonical pathway shares components with the other BMP signaling pathways, the balance among different BMP pathways may coordinate the NMJ development and function (Sulkowski, 2016).

The complexity of BMP signaling at the Drosophila NMJ is reminiscent of the neurotrophin-regulated signaling in vertebrate systems. Neurotrophins were first identified as neuronal survival factors. Like BMPs, they are secreted as pro-proteins that must be processed to form mature ligands. The active dimers bind to transmembrane kinase receptors and induce their activation through trans-phosphorylation. Neurotrophin/receptor complexes are internalized and transported along axons to the cell soma; signaling in the cell soma controls gene expression and promotes neuronal differentiation and growth. In addition, local neurotrophin signaling regulates growth cone motility, enhances the presynaptic release of neurotransmitter and mediates activity-dependent synapse formation and maturation. At the Drosophila NMJ, several neurotrophins have been implicated in neuron survival, axon guidance and synapse growth. It will be interesting to test for the crosstalk between neurotrophin and BMP signaling at these synapses (Sulkowski, 2016).

The novel noncanonical BMP pathway reported in this study is the first example of a BMP pathway triggered by selective neurotransmitter receptors and influencing receptor distribution at PSDs. It is expected that some of these functions will apply to mammalian glutamatergic synapses: First, as indicated in the Allen Brain Atlas, glutamate receptors and Neto proteins are widely expressed in mammalian brain structures where BMPs, BMPRs and Smads are expressed. Second, BMPs have been shown to rapidly potentiate glutamate-mediated currents in human retina neurons, presumably via a noncanonical pathway. Finally, mice lacking Chordin, a BMP antagonist, have enhanced paired-pulse facilitation and LTP and show improved learning in a water maze test. Such changes could not be explained by Smad-dependent transcriptional responses and were not accompanied by structural alterations in synapse morphology. Instead, presynaptic noncanonical BMP pathway may influence the activity of postsynaptic glutamate receptors by modulating their synaptic distribution and stability (Sulkowski, 2016).


PROTEIN STRUCTURE

Amino Acids - 570

Structural Domains

saxophone was cloned on the basis of its containing homology to mammalian serine-threonine kinases. Degenerate primers were designed in order to isolate candidate receptors by polymerase chain reaction. The primers corresponded to the conserved cytoplasmic domains of the serine-threonine kinase receptors of activin and TGF-beta (Xie, 1994).

The serine-threonine kinase domains of the Drosophila and vertebrate receptors are 78% homologous, the two fly genes (sax and tkv) being no more closely related to one another than they are to their vertebrate homologs. The extracellular domains show resemblence only in the spacing of the cysteine residues (Nellen, 1994 and Xie, 1994).


EVOLUTIONARY HOMOLOGS

Two structural elements, the L45 loop on the kinase domain of the transforming growth factor-beta (TGF-beta) family type I receptors and the L3 loop on the MH2 domain of Smad proteins, determine the specificity of the interactions between these receptors and Smad proteins. The L45 sequence of the TGF-beta type I receptor (TbetaR-I) specifies Smad2 interaction, whereas the related L45 sequence of the bone morphogenetic protein (BMP) type I receptor (BMPR-I) specifies Smad1 interactions. Members of a third receptor group, which includes ALK1 and ALK2 from vertebrates and Saxophone from Drosophila, specifically phosphorylate and activate Smad1 even though the L45 sequence of this group is very divergent from that of BMPR-I. The structural elements that determine the specific recognition of Smad1 by ALK1 and ALK2 have been investigated. In addition to the receptor L45 loop and the Smad1 L3 loop, the specificity of this recognition requires the alpha-helix 1 of Smad1. The alpha-helix 1 is a conserved structural element located in the vicinity of the L3 loop on the surface of the Smad MH2 domain. Thus, Smad1 recognizes two distinct groups of receptors (the BMPR-I group and the ALK1 group) through different L45 sequences on the receptor kinase domain and a differential use of two surface structures on the Smad1 MH2 domain (Chen, 1999).

TGFbeta signaling pathways of the bone morphogenetic protein (BMP) subclass are essential for dorsoventral pattern formation of both vertebrate and invertebrate embryos. It has been determined, by chromosomal mapping, linkage analysis, cDNA sequencing and mRNA rescue, that the dorsalized zebrafish mutant lost-a-fin (laf) is defective in the gene activin receptor-like kinase 8 (alk8), which encodes a novel type I TGFbeta receptor. The alk8 mRNA is expressed both maternally and zygotically. Embyros that lack zygotic, but retain maternal Laf/Alk8 activity, display a weak dorsalization restricted to the tail and die by 3 days postfertilization. The laf dorsalized mutant phenotype was rescued by alk8 mRNA injection. Homozygous laf/alk8 mothers were generated to investigate the maternal role of Laf/Alk8 activity. Adult fish lacking Laf/Alk8 activity are fertile, exhibit a growth defect and are significantly smaller than their siblings. Embryos derived from homozygous females, which lack both maternal and zygotic Laf/Alk8 activity, display a strongly dorsalized mutant phenotype, no longer limited to the tail. These mutant embryos lack almost all gastrula ventral cell fates, with a concomitant expansion of dorsal cell types. During later stages, most of the somitic mesoderm and neural tissue circumscribe the dorsoventral axis of the embryo. Zygotic laf/alk8 mutants can be rescued by overexpression of the BMP signal transducer Smad5, but not the Bmp2b or Bmp7 ligands, consistent with the Laf/Alk8 receptor acting within a BMP signaling pathway, downstream of a Bmp2b/Bmp7 signal. Antibodies specific for the phosphorylated, activated form of Smad1/5, show that BMP signaling is nearly absent in gastrula lacking both maternal and zygotic Laf/Alk8 activity, providing further evidence that Laf/Alk8 transduces a BMP signal. In total, this work strongly supports the role of Laf/Alk8 as a type I BMP receptor required for the specification of ventral cell fates (Mintzer, 2001).

In Drosophila, two BMP type I receptors, Thickveins and Saxophone, act in the transduction of the Dpp and Screw signals, respectively. However, Dpp and Screw together with their respective receptors play non-equivalent roles in dorsoventral pattern formation, unlike the Bmp2b and Bmp7 ligands in the zebrafish. Thickveins and Dpp act in the establishment of all dorsal cell fates, while Saxophone and Screw specify only a subset. The Laf/Alk8 receptor, which is more similar to Saxophone phlyogenetically than Thickveins, plays an extensive role in dorsoventral pattern formation, more similar to Thickveins in the fly. It is possible that all Bmp2b and/or Bmp7 signaling acts through the Laf/Alk8 type I receptor in dorsoventral patterning; however, this does not exclude the role of additional type I receptors in this process. In the fly, Thickveins, Saxophone, and two Punt type II receptor subunits are proposed to form a tetrameric complex together with one Dpp and one Screw homodimer. Hence, additional type I receptors may function together with the Alk8/Laf receptor. Further studies are required to determine the roles of other BMP type I receptors in dorsoventral patterning in vertebrates (Mintzer, 2001).

Neural crest cells (NCCs) are pluripotent migratory cells that contribute to the development of various craniofacial structures. Many signaling molecules have been implicated in the formation, migration and differentiation of NCCs, including bone morphogenetic proteins (BMPs). BMPs signal through a receptor complex composed of type I and type II receptors. Type I receptors (Alk2, Alk3 and Alk6) are the primary determinants of signaling specificity and therefore understanding their function is important in revealing the developmental roles of molecular pathways regulated by BMPs. A Cre/loxP system has been used for neural crest specific deletion of Alk2. The results show that mice lacking Alk2 in the neural crest display multiple craniofacial defects, including cleft palate and a hypotrophic mandible. It is concluded that signaling via Alk2 receptors is non-redundant and regulates normal development of a restricted set of structures derived from the cranial neural crest (Dudas, 2004).

Cardiac neural crest cells are multipotent migratory cells that contribute to the formation of the cardiac outflow tract and pharyngeal arch arteries. Neural crest-related developmental defects account for a large proportion of congenital heart disorders. Recently, the genetic bases for some of these disorders have been elucidated, and signaling pathways required for induction, migration and differentiation of cardiac neural crest have emerged. Bone morphogenetic proteins comprise a family of secreted ligands implicated in numerous aspects of organogenesis, including heart and neural crest development. However, it has remained generally unclear whether BMP ligands act directly on neural crest or cardiac myocytes during cardiac morphogenesis, or function indirectly by activating other cell types. Studies on BMP receptor signaling during organogenesis have been hampered by the fact that receptor knockouts often lead to early embryonic lethality. A Cre/loxP system was used for neural crest-specific deletion of the type I receptor, ALK2, in mouse embryos. Mutant mice display cardiovascular defects, including persistent truncus arteriosus, and abnormal maturation of the aortic arch reminiscent of common forms of human congenital heart disease. Migration of mutant neural crest cells to the outflow tract is impaired, and differentiation to smooth muscle around aortic arch arteries is deficient. Moreover, in Alk2 mutants, the distal outflow tract fails to express Msx1, one of the major effectors of BMP signaling. Thus, the type I BMP receptor ALK2 plays an essential cell-autonomous role in the development of the cardiac outflow tract and aortic arch derivatives (Kaartinen, 2004).


saxophone: Regulation | Developmental Biology | Effects of Mutation | References

date revised: 22 February 2022

Home page: The Interactive Fly © 1995, 1996 Thomas B. Brody, Ph.D.

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