Focal adhesion kinase-like


DEVELOPMENTAL BIOLOGY

Expression of Fak56D protein was examined throughout development. Total lysates from embryonic, first instar, third instar, early pupa, mid/late pupa, and adult developmental stages were analyzed on SDS-PAGE followed by immunoblotting for Fak56D. Levels of Fak56D expression are high during embryonic development, decrease early in larval development, and then are high again at later larval and pupal stages, indicating that Fak56D protein expression is indeed regulated during development. Interestingly, Fak56D appears as a doublet at all stages, although during the later stages of development the upper band is prevalent. Currently, the nature of this doublet is unknown; it may reflect the phosphorylation status of Fak56D, although alternative splicing could also be responsible, because this has previously been reported for both FAK and Pyk2. A 4.5-kb Fak56D RNA was detected by Northern blot analysis, and the levels of this RNA parallel the levels of Fak56D protein during development (Palmer, 1999).

Immunostaining of Drosophila embryos with affinity-purified anti-Fak56D antibodies shows strong Fak56D expression in the central nervous system. Immunostaining of primary cell cultures plated on laminin and tiggrin show that anti-Fak56D antibodies also strongly stain neurite networks. These data are consistent with in situ hybridization analysis of Fak56D RNA during development, which indicates that Fak56D is widely expressed during embryogenesis with a high level of expression within the developing nervous system. Additionally, in situ hybridization analysis of Drosophila embryos by the Berkeley Drosophila Genome Project using the CK1820 EST has revealed expression of Fak56D in the central nervous system, embryonic brain, epidermis, nerve cord, and visceral mesoderm, correlating with the in situ hybridization and immunostaining data (Palmer, 1999).

In stage 16 embryos, strong expression of the Fak56D mRNA is detected in the central nervous system and the junction of muscle and epidermis. Immunohistochemical analysis also showed expression of the Fak56D protein in the muscle attachment. A cross-sectioned view of the immunostained image suggests that Dfak56 is expressed predominantly in the epidermal cells, but less or not at all in the muscular cells (Fujimoto, 1999).

The subcellular localization of the Fak56D protein was examined using the Drosophila neuronal cell line BG2-c6. The BG2-c6 cells express the FAK56D mRNA. They also express integrins, and formation of integrin clusters is observed in cells with several focal adhesion proteins, including p21-activated kinase and alpha-actinin. Immunofluorescent staining of the BG2-c6 cells with anti-Fak56D antibodies produces a focal adhesion-like pattern. The nucleus is also stained with these antibodies. In some populations, the edges of the cells are also stained. Staining with anti-phosphotyrosine antibody produces a pattern that overlaps with that of anti-Fak56d antibodies, except for the nucleus. Staining with anti-phosphotyrosine and anti-betaPS integrin antibodies shows colocalization of tyrosine-phosphorylated proteins with integrin. These data suggest that Fak56D is a component of focal adhesions in Drosophila (Fujimoto, 1999).

To determine when Fak56D is expressed during development, a developmental Northern blot was carried out with a probe from the 5' end of the Fak56D cDNA: three bands of 4.7, 5.3, and 6.5 kb were detected. The 4.7- and 5.3-kb transcripts are expressed during all developmental stages examined, but most notably during embryogenesis. During larval and pupal stages, expression is reduced. Expression is high in ovaries. The abundance of these messages in both 0- to 2-hr embryos and ovaries suggests a maternal contribution. The 6.5-kb message is present at comparatively low levels during development but is up-regulated at 8- to 14-hr of embryogenesis. Identical results were obtained with a Fak56D 3'UTR probe (Fox, 1999).

In situ hybridization to Drosophila embryos was performed by using a biotinylated anti-sense RNA probe. At early stages of development, Fak56D is expressed throughout the embryo. After cellularization, there is an abrupt, yet transient, drop in the level of mRNA in the blastoderm. Expression levels increase during gastrulation, with highest levels in the mesoderm. At stage 13, Fak56D mRNA decreases in some cells, giving rise to a pattern of segmental stripes, and enriched expression in the gut and central nervous system becomes apparent. By stage 14, the expression pattern is once again uniform, with the exception of elevated expression in the brain (Fox, 1999).

Guinea pig antibodies were raised against the C-terminal domain of Fak56D and embryos were stained. Consistent with the mRNA expression pattern, Fak56D protein is uniformly distributed during early embryogenesis, although there is no drop in protein levels after cellularization of the blastoderm. Expression increases in the central nervous system and gut during stage 13, and these heightened levels persist through stage 15 and beyond. Variation in protein concentration along the length of the body wall first appears at stage 15. Examination of the body wall from stage 16 embryos at higher magnification shows that the antiserum does not detect accumulation of Fak56D protein in muscle attachment sites, although there is a striking decrease in signal in ectodermal cells located at the segmental boundaries (Fox, 1999).

Given the presence of Fak56D mRNA in ovaries, developing egg chambers were stained with Fak56D antiserum. Fak56D is abundant in the germ cells at early stages of oogenesis, but decreases significantly by stage 6. A similar, but less dramatic, decrease occurs in the somatic follicle cells, but they continue to express Fak56D at later stages. Strikingly, the level of Fak56D does not drop in follicle cells at the anterior end of stage 6 egg chambers. An enlarged view of the anterior tip of the stage 6 egg chamber shows clearly the difference in Fak56D levels between follicle cells at the anterior tip and those more posterior. This elevated level of Fak56D protein persists through early stage 9, when a group of follicle cells, known as the border cells, begin to migrate between the nurse cells toward the oocyte. The border cells originate from the anterior tip of the egg chamber and thus contain high levels of Fak56D protein. Fak56D is prominent in the border cells throughout their migration. Notably, Fak56D does not localize to ring canals in which elevated levels of phosphotyrosine and F-actin have been reported. It does, however, seem to accumulate in the basal region of the posterior follicle cells (Fox, 1999).

The Drosophila gene taiman encodes a steroid hormone receptor coactivator related to AIB1. Mutations in tai cause defects in the migration of specific follicle cells, the border cells, in the Drosophila ovary. Drosophila E-cadherin (Shotgun) is required for border cell migration. To determine whether the tai migration defect might be due to reduction in Shotgun expression, egg chambers containing tai mutant clones were stained with antibodies against Shotgun. In all wild-type stages examined, Shotgun accumulates in the central, nonmigratory polar cells, as well as in the junctions between individual border cells. Shotgun colocalizes with cortical F-actin in these locations. Prior to migration, when the border cells are still part of the follicular epithelium, Shotgun also accumulates at the junctions between border cells and nurse cells. However, once the border cells leave the follicular epithelium and invade the neighboring germline cell cluster, much less Shotgun staining is evident at the junctions between the nurse cells and border cells, relative to the level between border cells or in the polar cells. When migration is complete, Shotgun accumulates again in the junctions between the border cells and the oocyte (Bai, 2000).

In tai mutant clusters, Shotgun staining is abnormally elevated at the border cell/nurse cell junctions. In contrast, in slbo mutants, Shotgun expression fails to rise at the time of migration and Shotgun immunoreactivity is only detected at high levels within the polar cells. Armadillo (Arm) colocalizes with Shotgun in wild-type and mutant border cells. The abnormal accumulation of Shotgun and Arm in tai mutants does not appear to result from increased transcription of Shotgun because overexpression of Shotgun in border cells causes neither a migration defect nor specific accumulation of cadherin staining at the border cell/nurse cell junctions. Nor does the abnormal accumulation of Shotgun and Arm appear to be simply a consequence of the migration failure. In addition to slbo, Shotgun and Arm expression were examined in border cells that fail to migrate due to mutations in the jing locus: no defect in either expression or localization of adhesion complexes was observed. Nor are defects in either Shotgun or Arm expression or localization found in border cells that fail to migrate due to expression of dominant-negative Rac (Bai, 2000).

The accumulation of Shotgun at the border cell/nurse cell boundary suggests that the role of tai in border cell migration might be to stimulate turnover of adhesion complexes during migration in order to allow forward movement. One protein believed to play a role in turnover of adhesion complexes is Focal adhesion kinase. Drosophila FAK (Fak56D) is highly enriched in the border cells during their migration, but not in the polar cells (Bai, 2000).

To determine whether Fak56D expression or localization is affected by mutations that disrupt border cell migration, wild-type and slbo mutant egg chambers were stained and the staining was compared to that of egg chambers containing tai mosaic clones. Fak56D expression is significantly reduced in slbo mutant border cells. Furthermore, the level of reduction correlates with the degree of inhibition of migration. That is, in some slbo egg chambers, border cell migration fails completely and the cells remain at the anterior tip. In such egg chambers, Fak56D expression is undetectable. In a minority of slbo mutant chambers, the cells migrate a little. In these egg chambers, Fak56D expression is reduced compared to wild type, but is detectable. In tai mutant border cells, Fak56D expression is present; however, its distribution is altered relative to wild type. Rather than being evenly distributed throughout the cytoplasm, Fak56D appears to accumulate at the would-be leading edge of the cluster. Some border cell clusters that are mutant for tai exhibit partial migration and in these clusters, the abnormal distribution of Fak56D is only slightly affected such that little Fak56D accumulation can be detected at the most posterior position within the cluster. Thus, the severity of the migration defect in tai mutants correlates with the severity of the defect in Fak56D localization (Bai, 2000).

Effects of Mutation

The mammalian focal adhesion kinase (FAK) family of non-receptor protein-tyrosine kinases has been implicated in controlling a multitude of cellular responses to the engagement of cell-surface integrins and G-protein-coupled receptors. The high level of sequence conservation between the mammalian proteins and the Drosophila homologue of FAK, Fak56, suggests that Fak56 would have similar functions. However, Drosophila Fak56 is shown to be not essential for integrin functions in adhesion, migration or signaling in vivo. Furthermore, animals lacking Fak56 are viable and fertile, demonstrating that Fak56 is not essential for other developmental or physiological functions. Despite this, overexpressed Fak56 is a potent inhibitor of integrins binding to the extracellular matrix, suggesting that Fak56 may play a subtle role in the negative regulation of integrin adhesion (Grabbe, 2004).

While the conclusion that FAK family PTKs are not required in Drosophila, and are thus are not essential players in integrin function in vivo, data from C. elegans indicate that FAK is also non-essential in the nematode. There appears to be only one FAK family member in the genome of this species, thus eliminating the possibility of redundancy explaining these results. While this conclusion has not been formally reported for C. elegans, two separate findings support this hypothesis: (1) RNAi towards FAK (known as kin-32 in the nematode) causes no obvious phenotypes, and (2) a large deletion in the kin-32 open reading frame is viable (Grabbe, 2004).

In Drosophila, mutants for many of the proteins known to localize at the muscle attachment site cause lethality, in many cases due to muscle detachment, i.e. the integrins themselves, ILK, Talin, Tiggrin (although 1% of flies eclose), Laminin, alpha-actinin, and PINCH. Other molecules, such as the Drosophila Tensin homologue (also known as Blistery), are not lethal, but do display phenotypes indicative of failure of adhesion, such as wing blistering. However, it is interesting to note that another cytoskeletal protein -- Vinculin -- has also been shown to be non-essential in Drosophila, in contrast with previous results in the mouse, where animals mutant for the vinculin gene display a lethal phenotype due to heart and brain defects during embryogenesis. The non-essential nature of Fak56 in Drosophila perhaps explains why extensive attempts to target Fak56 through various methods have thus far been unsuccessful, since the general assumption has been that such mutants would be lethal. Moreover, this may also be the reason why, despite extensive genetic screening by several groups with the purpose of identifying molecules involved in integrin-mediated signaling, no mutations have ever been identified in Fak56, although the effectiveness of these screens has been demonstrated by the fact that they have independently identified several common loci in addition to existing PS integrin genes (Grabbe, 2004).

While Fak56 protein appears to be ubiquitously expressed, phospho-Fak56 is strongly localized at muscle attachment sites. This implies that Fak56 is not only localized, but also activated at these locations, since phosphorylation of the FAKY397 site (which is conserved in Fak56), is considered to reflect FAK activation in vivo. The anti-phospho-FAKY397 antibodies seem to be specific for phosphorylated Drosophila Fak56, based on two criteria: (1) loss of immunoreactivity in the Fak56 mutants, and (2) overexpressed wild-type Fak56 is recognized by the antiphospho-FAKY397 antibodies, whereas overexpressed Fak56Y430F mutant protein is not. A role for Fak56, albeit an accessory one, at muscle attachment sites, is endorsed by the finding that phosphorylated Fak56 is absent from muscle attachment sites in integrin mutants. Thus, while not required for integrin function, Fak56 appears to be involved through an as yet undefined mechanism in integrin-mediated events in Drosophila embryogenesis. In spite of the absence of the Fak56 PTK at muscle attachment sites in Fak56 mutant embryos, the levels of phosphotyrosine observed are indistinguishable from wild-type, suggesting that another PTK(s) is activated at these sites. This is interesting in light of a recent report that Src kinases can be activated by direct interaction with integrin ß-subunit tails. Such a model implies that Src family kinases can be activated by integrins independently of FAK and could explain the unexpected lack of phenotypes in the Fak56 mutant. Interestingly, it has recently been shown that v-Src transformation of Ptk2-/- fibroblasts rescues the motility defects observed in Ptk2-/- fibroblasts, which is in keeping with these results (Grabbe, 2004).

Consistent with an accessory involvement of FAK in integrin-mediated adhesion, the Fak56-induced wing blister phenotype is sensitized in an integrin mutant background, thus indicating a genetic interaction between Fak56 and integrins in this context. Employing the UAS-GAL4 system to drive Fak56 specifically in muscles it is clear that overexpression of Fak56 disrupts muscle attachment, thus overexpression of Fak56 causes much more serious defects than its absence. This is consistent with Fak56 either functioning as an adaptor by displacing more critical proteins from the integrin cytoplasmic tail required for the extracellular binding to ECM ligands, or causing a dissociation of the integrin-containing complex by excessive phosphorylation. The evidence in mammalian systems suggests that FAK plays a critical role in cell migration, which is a complex, highly regulated process that involves the continuous formation and disassembly of adhesions. Indeed, recently it has elegantly been shown that FAK is important for adhesion turnover at the cell front, a process central to migration. The migration of (1) the endoderm, (2) border cells during oogenesis, (3) germ cells and (4) the trachea, has been examined in Fak56 mutant animals, and no defects were found in any process in the absence of Fak56. However, such a role of FAK in the disassembly, rather than in the actual assembly of these structures, is in agreement with findings that Fak56 overexpression results in detached muscles where the alphaPS2 integrin is still localized at the muscle ends, as though dissociated from the ECM. This then raises the question of why Fak56 is normally found in the developing muscles. It is speculated that Fak56 contributes to keeping the strong adhesive junctions dynamic so that they can be remodeled during the formation of the attachments and the subsequent growth of the muscles. Perhaps a defect in this process would be more apparent under more strenuous growth conditions than those found in the laboratory (Grabbe, 2004).

One important point to consider is the wealth of data from mammalian cell experiments indicating a critical role for FAK family PTKs in integrin function. Such a function is corroborated by the phenotype of the FAK knockout mouse, which dies early in embryogenesis. Indeed, Ptk2-/- fibroblasts derived from FAK mutant embryos, exhibit a rounded morphology, and have an increased number of focal contact sites and decreased rates of cell migration. A recent RNAi-based screen of the Drosophila genome to find novel genes affecting cell morphology identified Fak56 as a molecule affecting cell spreading. In addition, overexpression of Fak56 in the fly using the GAL4-UAS system also produces a wealth of integrin-related phenotypes. Overexpressed Fak56 does localize to muscle attachment sites under such conditions, and it is entirely possible that this creates a neomorphic phenotype, i.e. such that the presence of excess Fak56 protein -- or indeed Fak56 activity -- binds up multiple factors and acts in a dominant negative fashion to produce these defects. Thus, experiments in mammalian cells employing overexpression of FAK to analyze integrin function may not be an ideal model for the in vivo scenario. Furthermore, a radical explanation for the strong defects observed in Ptk2-/- fibroblasts, which would be consistent with the current findings, is if the primary defect of removing FAK is the observed elevated expression and activity of the related kinase Pyk2, and the Src family PTKs. If overexpressed Pyk2 causes similar dominant negative effects on other components of integrin adhesion, as is seen when Fak56 is overexpressed, then this could contribute to the severe phenotype observed in the FAK mutant mice. Finally, it is also possible that the functions of the FAK family PTKs are not totally conserved between Drosophila and mice; this suggests that FAK has assumed a more critical role during evolution of vertebrates (Grabbe, 2004).

Disruption of the single Drosophila Fak56 gene is the first example of an animal completely lacking FAK PTK function. However, it is surprising that a genetic null for a protein such as FAK, that has always been thought to be a critical link between integrins and the actin cytoskeleton, exhibits a viable phenotype. Since this also appears to be true for the C. elegans FAK family PTK, it must be assumed that while FAK may play an accessory role in modulating integrin functions in vivo, it is by no means essential for integrin-mediated adhesion or signaling in the fruitfly. It is obvious that further experiments will be required to define and understand the exact role of the Fak56 PTK in such processes in vivo (Grabbe, 2004).

Focal adhesion kinase controls morphogenesis of the Drosophila optic stalk

Photoreceptor cell axons (R axons) innervate optic ganglia in the Drosophila brain through the tubular optic stalk. This structure consists of surface glia (SG) and forms independently of R axon projection. A screen for genes involved in optic stalk formation identified Fak56D, encoding a Drosophila homolog of mammalian focal adhesion kinase (FAK). FAK is a main component of the focal adhesion signaling that regulates various cellular events, including cell migration and morphology. Fak56D mutation causes severe disruption of the optic stalk structure. These phenotypes were completely rescued by Fak56D transgene expression in the SG cells but not in photoreceptor cells. Moreover, Fak56D genetically interacts with myospheroid, which encodes an integrin ß subunit. In addition, CdGAPr is also required for optic stalk formation and genetically interacts with Fak56D. CdGAPr encodes a GTPase-activating domain that is homologous to that of mammalian CdGAP, which functions in focal adhesion signaling. Hence the optic stalk is a simple monolayered structure that can serve as an ideal system for studying glial cell morphogenesis and the developmental role(s) of focal adhesion signaling (Murakami, 2007).

The Drosophila visual system consists of the compound eyes and the optic ganglia. The compound eye consists of approximately 750 ommatidial units, each of which contains eight photoreceptor cells. During the development of the visual system, photoreceptor cells send their axons (R axons) from the eye primordium (eye disc) to their targets in the optic lobe of the brain through a structure called the optic stalk. All R axons from an eye disc come together and make a single thick bundle in the optic stalk. The optic stalk also contains two kinds of glial cells, the wrapping glial (WG) cells and surface glial (SG) cells. The WG cells are intermingled with R axons and extend long processes that wrap around several R axons, so that the entire R axon bundle is subdivided into groups of smaller bundles. The WG processes form an inner sheath to segregate these bundles from each other during the pupal stage. By contrast, SG cells form an outer sheath that surrounds the entire bundle of R axons. It is clear that the optic stalk glia play roles in R axon innervation and ensheathment; however, the precise structure and development of the optic stalk remain largely unknown (Murakami, 2007).

SG cells in the optic stalk have characteristic bipolar morphology and form a single-cell monolayer covered by a basement membrane (BM). Optic stalk expansion occurs before R axon innervation. Moreover, even in those mutants with no R axon innervation, SG cells form a single-cell-layer tube that develops normally. Therefore, it is concludes that SG cells autonomously form the optic stalk. This study sought to elucidate the molecular mechanisms underlying optic stalk formation. In screening for mutants that exhibit defects in R axon innervation or for genes that are specifically expressed in the optic stalk, the Fak56D and CdGAPr genes were identified as encoding important components required for optic stalk formation. Fak56D is a Drosophila homolog of mammalian focal adhesion kinase (FAK; also known as Ptk2), which is known to be a main regulatory component of focal contacts. Focal contacts are large integrin complexes that anchor the cytoskeleton of cells to the extracellular matrix. While focal contacts generate traction forces during migration, their disassembly is also crucial to the control of cell migration. FAK is involved in cell migration through disassembly of focal contacts, or through regulation of cytoskeletal rearrangement via Rho GTPases. In Drosophila, several studies indicate that Fak56D also acts in focal contacts in a similar way to that of mammalian FAK. Fak56D is tyrosine-phosphorylated and localized to focal contacts upon the plating of embryonic cells onto extracellular matrix components. However, as no Fak56D loss-of-function phenotype has been reported, the in vivo function of Fak56D remains elusive (Murakami, 2007 and references therein).

In Fak56D mutant animals, SG cells were not arranged into a tubular structure, although cell proliferation and differentiation were normal. Clone labeling analysis suggested that SG cell distribution along the anteroposterior (AP) axis was defective in Fak56D mutants. This study also identified CdGAPr as a possible functional partner of Fak56D. CdGAPr has a GTPase activating domain that is homologous to that of mammalian CdGAP (Sagnier, 2000). It has been shown that mammalian CdGAP regulates cell cytoskeletal rearrangement through Rac or Cdc42; however, in vivo function of neither CdGAP nor CdGAPr has been described. This study found that loss-of-function alleles of CdGAPr exhibited a Fak56D-like phenotype. Moreover, a strong genetic interaction was observed between Fak56D and CdGAPr, indicating that these genes act together to regulate SG cell behavior. Recently, mammalian CdGAP was shown to localize to focal contacts, and to be required for regulation of cell morphology and motility (LaLonde, 2006). The current results provide strong evidence that optic stalk shape is controlled by mechanisms acting autonomously in SG cells, in which Fak56D and CdGAPr play crucial roles (Murakami, 2007).

SG cells are regularly arranged and form the optic stalk, a monolayered tube that surrounds the entire R axon bundle. During larval development, the optic stalk becomes larger as SG cells proliferate, and this seems to adjust the size of the stalk so that it can wrap around the R axon bundle. The proliferation and morphogenesis of SG cells could depend on signals from the incoming R axons, because various cellular events of glial cells, such as differentiation, migration and proliferation, have been shown to require interaction with neuronal axons. For example, in the developing rodent optic nerve, Sonic hedgehog (Shh) from retinal ganglion cells (RGCs) promotes the proliferation of astrocytes. Moreover, initial formation of the optic stalk during embryogenesis was shown to be dependent on the axons from larval photoreceptor cells. However, the current data demonstrate that SG cells in Drosophila form the optic stalk independently of R axons. (1) SG cells proliferate before R axon innervation, which leads to the expansion of the optic stalk. (2) In so1 mutants the optic stalk develops normally despite complete lack of R axon innervation. In addition, specific expression of a Fak56D transgene in SG but not photoreceptor cells rescued a defect in optic stalk morphology, hence indicating the presence of an intrinsic mechanism in SG cells to regulate optic stalk morphogenesis. Some kinds of glial cells are reported to develop independently of axon innervation in a similar way to SG cells. For instance, during Drosophila visual system development, retinal basal glia (RBG) are derived from the optic stalk and migrate into the eye disc. This process was shown to be independent of R axons. In these cases, glial cells must behave by unknown mechanisms independent of axon cues. Since the current results demonstrated that SG cell development is highly independent of R axons, this system can provide an excellent model to elucidate mechanisms underlying axon-independent glial development and behavior (Murakami, 2007).

This study has elucidated cellular mechanisms underlying optic stalk morphogenesis in Drosophila. Optic stalk morphology and development are precisely regulated, and the optic stalk expands throughout the larval stages via cell proliferation. SG cells are distributed along the AP axis during optic stalk formation. Since cells in a clone are often associated with each other, SG cells may migrate along the nearby SG cells that have extended processes along the AP axis. Such homotypic migration is reported for neuronal precursors in the adult mammalian subventricular zone (SVZ). Neuronal precursors migrate to the olfactory bulb in chains, by sliding along each other without the assistance of other cell types. Such directed distribution of SG cells is likely to be necessary for keeping the optic stalk thin and long. The mechanism regulating tube morphogenesis is largely unknown. The finding that SG cells undergo cell arrangement during optic stalk expansion provides an interesting insight into the morphogenesis of a tubular structure (Murakami, 2007).

It is known that the optic stalk disappears during the pupal stage and that the ommatidia are set much closer to the optic lobe. This study found that SG cells begin to locate at the surface of the optic lobe during early pupal stages. This is similar to what was observed in Fak56D mutants during larval stages. It seems that the optic stalk is degenerating during the pupal stage through relocation of SG cells to the optic lobe, and this process might be regulated by FAK activity (Murakami, 2007).

In an attempt to identify the molecular mechanisms underlying optic stalk formation, it was found that the optic stalk was disrupted in Fak56D mutants. Expression of a Fak56D transgene in SG cells, but not photoreceptor cells, rescued the defect. This indicates that Fak56D is autonomously required in SG cells. Fak56D is apparently required during the expansion of the optic stalk. Since differences were detected in number of SG cells between wild type and Fak56D mutants, it is unlikely that Fak56D regulates the proliferation of SG cells. SG cells are mis-localized on the surface of the optic lobe in Fak56D mutants instead of forming a tubular structure. Visualization of newly divided cells by clonal labeling suggests that SG cells tend to migrate along the AP axis, and this could lead to formation of a longitudinal tube structure. In Fak56D mutants SG cells partially lose tendency to migrate along the AP axis; this could lead to enlargement of the diameter as opposed to the length. Ectopic localization of SG cells on the optic lobe observed in Fak56D mutants is likely to be the result of the optic stalk widening. Mammalian FAK plays a central role in cell migration; hence in Drosophila Fak56D may regulate optic stalk formation via regulation of cell migration (Murakami, 2007).

Fak56D might also be required for adhesion between SG cells. siRNA-mediated mammalian FAK knockdown results in loss of N-cadherin-based cell-cell adhesion in Hela cells. In addition, it has been shown that overexpression of a kinase-defective mutant of FAK in cultured cells blocks the hyperosmolarity-induced E-cadherin accumulation at the cell periphery. Since SG cells are closely attached to each other, adhesion between SG cells may be important for keeping optic stalk tubular structure. SG cells are also attached to the BM throughout optic stalk formation. Because Fak56D is implied to regulate integrin adhesion, it is possible that Fak56D is required for SG cells to form proper adhesion to the BM (Murakami, 2007).

In addition to Fak56D, CdGAPr was identified as a regulator of optic stalk morphology. Fak56D and CdGAPr exhibit a strong genetic interaction. Since both mammalian FAK and CdGAPr are known to act at focal contacts, this raised the possibility that focal contacts are important for optic stalk morphogenesis. Main components of focal contacts, such as integrins or Paxillin, are conserved between vertebrates and invertebrates. In Drosophila, Fak56D is implicated in integrin-involving molecular mechanisms. This study found a genetic interaction between Fak56D and myospheroid (mys), which encodes a ß-subunit of integrin, ßPS. This suggests that Fak56D and CdGAPr act together with integrins in focal contacts to regulate SG cell behavior. Because the interaction between Fak56DCG1 and mys1 (a null allele) is much weaker than the interaction between Fak56DCG1 and CdGAPr hypomorphic alleles, it is possible that another ß subunit, ßnu, compensates for the loss of ßPS. ßPS and ßnu have shown to act together to regulate midgut cell migration. It is also possible that Fak56D function is regulated via other receptors, such as G-protein coupled receptors (GPCRs). Mammalian Pyk2, another FAK family member, is known to be activated via GPCRs (Murakami, 2007).

CdGAPr encodes a GAP domain that regulates the activity of Rho-family GTPases. The mammalian CdGAP regulates Rac and Cdc42, which are known regulators of the actin cytoskeleton, and are required for cell morphology and motility. This raises the possibility that FAK regulates cytoskeletal rearrangement via activation of CdGAP, although exact functional interaction between the two proteins remains elusive. In fact, the mammalian FAK interacts directly with guanine nucleotide exchange factors or GTPase-activating proteins in the regulation of cytoskeleton. More biochemical studies are required for elucidating the molecular mechanisms that underlie cell behaviors regulated by focal adhesion signaling (Murakami, 2007).

Dcas supports cell polarization and cell-cell adhesion complexes in development

Mammalian Cas proteins regulate cell migration, division and survival, and are often deregulated in cancer. However, the presence of four paralogous Cas family members in mammals (BCAR1/p130Cas, EFS/Sin1, NEDD9/HEF1/Cas-L, and CASS4/HEPL) has limited their analysis in development. The single Drosophila Cas gene, Dcas, was deleted to probe the developmental function of Dcas. Loss of Dcas had limited effect on embryonal development. However, Dcas is an important modulator of the severity of the developmental phenotypes of mutations affecting integrins (If and mew) and their downstream effectors Fak56D or Src42A. Strikingly, embryonic lethal Fak56D-Dcas double mutant embryos had extensive cell polarity defects, including mislocalization and reduced expression of E-cadherin. Further genetic analysis established that loss of Dcas modified the embryonal lethal phenotypes of embryos with mutations in E-cadherin (Shg) or its signaling partners p120- and beta-catenin (Arm). These results support an important role for Cas proteins in cell-cell adhesion signaling in development (Tikhmyanova, 2010).

This work identifies a strong interaction between the Dcas, and integrin pathway genes, including integrins and their effector kinases Fak56D and Src42A, during early embryonal development in Drosophila. The synthetic lethal phenotypes found in double mutants of Dcas and Src or FAK56D were marked by defects in dorsal closure and in some cases by the appearance of anterior cuticle holes that suggested head involution defects. These defects were commonly accompanied by abnormalities in epithelial function, including failure to appropriately localize shg/E-cadherin to cell junctions, and reduced shg expression. The data are compatible with the idea that either Fak56D or Dcas is sufficient to support shg/E-cadherin localization and cell polarization during morphogenetic movements in Drosophila embryos, but the absence of both cannot be sustained (Tikhmyanova, 2010).

Building from these observations, a novel synthetic lethal relationship was established between DCas, shg, and arm. As with crosses to alleles of Fak56D and Src42A, the point of lethality was at the time of dorsal closure, at embryonal stages 15-16, and associated with defective cuticle formation. One way to integrate these observations is to hypothesize that the DCas, Fak56D, and Shg protein products are normally in dynamic balance, with Dcas regulating Shg cycling. The fact that Crb and Dlg1, a mammalian homolog of Dlg, have been reported to support Shg localization to adherens junctions, suggests that Dcas/Fak56/Src42A specifically interact to support this cell polarity/cell junctional control system. In this context, it is suggestive that the Crb family protein CRB3 has been described as part of a complex including CRB3, Pals1, and PatJ that becomes tightly associated with Src kinase during reorganization of cell polarity. In the absence of DCas and Fak56D, Shg cannot localize properly; the moderately elevated levels of Shg proteins found in these embryos most likely arises as part of a cellular compensatory mechanism in response to decreased functional Shg signaling complexes. In further indirect support of the idea that this is a specific Dcas action, the fact that genetic interactions were not observed between Dcas1 and Aur or Dock indicates that Dcas does not promiscuously interact with other genetic lesions to reduce viability (Tikhmyanova, 2010).

A previous study demonstrated a role for Dcas in axonal guidance in the development of the nervous system of adult flies (Huang, 2007). That work analyzed the hypomorphic Dcas mutant allele DcasP1, and the small deficiency Df(3L)Exel6083, including Dcas and five adjacent genes, which were also used in this study. The earlier study focused exclusively on analyzing the contribution of Dcas to axonal guidance in late (stage 16/17) embryos: in that analysis, Dcas functioned similarly to integrins, and genetically interacted with integrins (if, mew, and mys) in regulating axon guidance and axonemal defasciculation. In this context, it is intriguing that the mammalian Cas family NEDD9 gene is abundant during neuronal development, has been proposed as a candidate locus for oral cleft defects in humans based on its chromosomal location near the OFC-1 locus. Together these findings raise the possibility that this specific Dcas paralog has a specific role in human neuronal migration and morphogenesis of the head. As with the current data using the new Dcas1 allele, homozygous deletion of Dcas in conjunction with integrins had moderate effect on viability of adult flies, although this work for the first time demonstrates an interaction between Dcas and if and mew, and also between Dcas and Src, in regulation of wing development (Tikhmyanova, 2010).

Generation of the first null allele of Dcas provides a useful new tool to study the role of this protein in Drosophila development. This work illuminates the evolutionary conservation of Dcas function within the integrin and receptor tyrosine kinase network, including FAK, Src, and integrins genes. The finding that a low percentage of embryos with mutant Dcas and all embryos with double mutations in Dcas and Fak56D, have perturbed localization of polarity markers, including Shg, indicates a novel function for Cas family in regulation of cell polarity. To date, the evidence directly connecting Cas proteins to a known mechanism for control of cell polarity is sparse. Although NEDD9 was in fact discovered in a functional genomics screen for cell cycle and polarity modifiers in budding yeast (leading to its designation as HEF1, Human Enhancer of Filamentation 1), the mechanism involved was not established, and given the great evolutionary distance involved, may not be relevant to a role in metazoans. Both BCAR1 and NEDD9 interact physically with proteins that influence cell polarity controls during pseudopod extension and other actin polarization processes: these include the GTP exchange factor AND-34 (Tikhmyanova, 2010).

The data in the present study indicating genetic interactions with cell-cell junction regulatory proteins Shg, Arm and p120-catenin may have considerable significance in the sphere of cancer research, as it implies that overexpression of Cas proteins may promote cancer progression by influencing the polarized movement of cells and influencing lateral attachments. The fact that one report has indicated interactions between BCAR1 and nephrocystins at cell-cell junctions in polarized epithelial cells implies that a potentially direct interaction of Cas proteins in these structures is conserved through mammals. However, given the known interactions of Cas proteins with FAK and SRC at focal adhesions, another possibility is that Cas may additionally or alternatively impact Shg function through indirect signaling emanating from these structures. Notably, it has been reported that NEDD9 overexpression induced by dioxin caused downregulation of E-cadherin, and it will be of great interest to study the consequences of overexpressing Dcas on Drosophila development. Consequences for loss of NEDD9 expression on E-cadherin expression or localization are not yet known. Resolving these questions will provide intriguing directions for future studies (Tikhmyanova, 2010).

Activity-dependent retrograde laminin A signaling regulates synapse growth at Drosophila neuromuscular junctions

Retrograde signals induced by synaptic activities are derived from postsynaptic cells to potentiate presynaptic properties, such as cytoskeletal dynamics, gene expression, and synaptic growth. However, it is not known whether activity-dependent retrograde signals can also depotentiate synaptic properties. This study shows that laminin A (LanA) functions as a retrograde signal to suppress synapse growth at Drosophila neuromuscular junctions (NMJs). The presynaptic integrin pathway consists of the integrin subunit βν and focal adhesion kinase 56 (Fak56), both of which are required to suppress crawling activity-dependent NMJ growth. LanA protein is localized in the synaptic cleft and only muscle-derived LanA is functional in modulating NMJ growth. The LanA level at NMJs is inversely correlated with NMJ size and regulated by larval crawling activity, synapse excitability, postsynaptic response, and anterograde Wnt/Wingless signaling, all of which modulate NMJ growth through LanA and βν. These data indicate that synaptic activities down-regulate levels of the retrograde signal LanA to promote NMJ growth (Tsai, 2012).

This study proposes a plasticity mechanism by which the synapse growth (or size) can be modulated by larva crawling and synaptic activities. These activities modulate LanA-integrin signaling that functions to constrain NMJ growth. This trans-synaptic signaling functions in a retrograde manner, which requires postsynaptic muscle-derived LanA and presynaptic integrin. The model suggests various activities modulate NMJ growth by regulating the LanA level and integrin signaling (Tsai, 2012).

Regulation of LanA levels at NMJs is the major mechanism underlying this synaptic structural plasticity. The LanA levels at NMJs are tightly coupled to several synaptic activities that are involved in synaptic structural plasticity at NMJs. Wg signaling in both pre- and postsynaptic compartments are shown to modify synaptic structure at Drosophila NMJs. The channel mutations para and eag Sh alter both synaptic potential and NMJ size. Finally, manipulation of postsynaptic responses by altering the GluRIIA and GluRIIB compositions also fine tunes synapse size and pFAK levels. Activities that promote NMJ growth also down-regulated LanA levels at NMJs. In contrast, NMJ growth suppression was accompanied with LanA accumulation, establishing an inverse correlation between the LanA level and the NMJ size. Importantly, manipulation of the gene dosage of LanA (or βν) could override these synaptic activities in NMJ growth regulation. This study also showed that LanA down-regulation at NMJs preceded synaptic structural remodeling induced by larval crawling, further supporting that LanA is a major mediator of these activities to modulate NMJ growth (Tsai, 2012).

Integrin signaling activities play important roles in synapse development and plasticity. In mammalian central synapses, various integrin subunits are important to transmit postsynaptic signaling in various plasticity models may function redundantly with βν to mediate integrin signaling. This study indicates a distinct presynaptic integrin pathway that is likely composed of βν and αPS3 (encoded by Volado), as suggested by their strong genetic interaction in NMJ growth. In response to postsynapse-secreted LanA signals, activation of the presynaptic integrin is transmitted through Fak56 activation. Interestingly, the signaling activity is rather local, limited by the range of LanA distribution, and shown by muscle 6-specific rescue, although this does not exclude the involvement of signaling to the nuclei of motor neurons. The presynaptic integrin/Fak56 signaling is in turn mediated by two downstream signaling activities. The activation of NF1/cAMP signaling, which suppressed NMJ overgrowth induced by crawling activity or βν mutation. The integrin/Fak56 pathway also suppresses Ras/MAPK signaling Tsai, 2008), as shown by diphospho-ERK (dpERK) accumulation and Fas2 reduction at NMJs in high crawling condition. These pathways have been shown to regulate cell adhesion and cytoskeletal organization, leading to the stabilization of synapses. The activity-dependent depletion of the LanA laminins in the synaptic cleft would allow the remodeling of synapses and further growth of NMJs (Tsai, 2012).

The activity-dependent structural plasticity is specific to the presynaptic integrin pathway. hiw mutants that show large NMJ size still retained the structural plasticity and constant pFAK levels at NMJs. Interestingly, LanA levels were increased in hiw mutants, in contrast to other NMJ overgrown mutants. Two nonmutually exclusive mechanisms can regulate activity-dependent LanA expressions at NMJs. First, within hours of activity induction, the LanA levels can be regulated at NMJs by putative ECM regulators such as matrix metalloproteinases. Second, transcription regulation of LanA can provide long-term changes of LanA levels at NMJs. Activity-triggered presynaptic Wg secretion promotes Wg receptor DFz2 activation on both post- and presynaptic compartments. The LanA level is regulated by the anterograde Wg signaling that is transduced through nuclear entry of the DFz2 intracellular domain and its transcription activity. However, LanA is unlikely to mediate all aspects of Wg signaling activity as overexpression of LanA in postsynapses suppressed ghost bouton formation, a hallmark in disrupting Wg signaling. Postsynaptic BMP/Gbb functions as a retrograde signal to activate presynaptic BMP type II receptor Wit in response to synaptic activity. With the lack of genetic interaction between BMP/Gbb and integrin signaling components, and constant levels of phosphorylated Mothers against dpp (pMad) in different crawling activities, it is proposed that both BMP/Gbb and LanA pathways can function in parallel by retrograde mechanisms to regulate NMJ growth (Tsai, 2012).


REFERENCES

Alrutz, M. A. and Isberg, R. R. (1998). Involvement of focal adhesion kinase in invasin-mediated uptake. Proc. Natl. Acad. Sci. 95(23): 13658-63. PubMed Citation: 9811856

Almeida, E. A. C., et al. (2000). Matrix survival signaling: From fibronectin via Focal adhesion kinase to c-Jun NH2-terminal kinase. J. Cell Biol. 149: 741-754. PubMed Citation: 10791986

Ashton, G. H., et al. (2010). Focal adhesion kinase is required for intestinal regeneration and tumorigenesis downstream of Wnt/c-Myc signaling. Dev. Cell 19(2): 259-69. PubMed Citation: 20708588

Asthagiri, A. R., et al. (1999). Quantitative relationship among integrin-ligand binding, adhesion, and signaling via focal adhesion kinase and extracellular signal-regulated kinase 2. J. Biol. Chem. 274(38): 27119-27. PubMed Citation: 10480927

Bai, J. Uehara, Y. and Montell, D. J. (2000). Regulation of invasive cell behavior by Taiman, a Drosophila protein related to AIB1, a steroid receptor coactivator amplified in breast cancer. Cell 103: 1047-1058. 11163181

Beggs, H. E., et al. (2003). FAK deficiency in cells contributing to the basal lamina results in cortical abnormalities resembling congenital muscular dystrophies. Neuron 40: 501-514. 14642275

Brabant, M. C., Fristrom, D., Bunch, T. A. and Brower, D. L. (1996). Distinct spatial and temporal functions for PS integrins during Drosophila wing morphogenesis. Development 122(10): 3307-17. PubMed Citation: 8898242

Burgaya, F., et al. (1997). Alternatively spliced focal adhesion kinase in rat brain with increased autophosphorylation activity. J. Biol. Chem. 272(45): 28720-28725. PubMed Citation: 9353341

Calalb, M. B., Polte, T. R. and Hanks, S. K. (1995). Tyrosine phosphorylation of focal adhesion kinase at sites in the catalytic domain regulates kinase activity: a role for Src family kinases. Mol. Cell. Biol. 15: 954-963. 7529876

Carragher, N. O., et al. (2003). A novel role for FAK as a protease-targeting adaptor protein: Regulation by p42 ERK and Src. Curr. Biol. 13: 1442-1450. 12932330

Casamassima, A. and Rozengurt, E. (1998). Insulin-like growth factor I stimulates tyrosine phosphorylation of p130(Cas), focal adhesion kinase, and paxillin. Role of phosphatidylinositol 3'-kinase and formation of a p130(Cas).Crk complex. J. Biol. Chem. 273(40): 26149-56. PubMed Citation: 9748296

Chan, P. C., et al. (1999). Suppression of ultraviolet irradiation-induced apoptosis by overexpression of focal adhesion kinase in Madin-Darby canine kidney cells. J. Biol. Chem. 274(38): 26901-6. PubMed Citation: 10480899

Chen, H.-C., et al. (1995). Interaction of focal adhesion kinase with cytoskeletal protein talin. J. Biol. Chem 270: 16995-16999. PubMed Citation: 7622520

Chen, H. C., et al. (1998). Tyrosine phosphorylation of focal adhesion kinase stimulated by hepatocyte growth factor leads to mitogen-activated protein kinase activation. J. Biol. Chem. 273(40): 25777-82. PubMed Citation: 9748249

Della Rocca, G. J., et al. (1999). Pleiotropic coupling of G protein-coupled receptors to the mitogen-activated protein kinase cascade. Role of focal adhesions and receptor tyrosine kinases. J. Biol. Chem. 274(20): 13978-84. PubMed Citation: 10318809

Fox, G. L., Rebay, I. and Hynes, R. O. (1999). Expression of DFak56, a drosophila homolog of vertebrate focal adhesion kinase, supports a role in cell migration in vivo. Proc. Natl. Acad. Sci. 96(26): 14978-83. PubMed Citation: 10611323

Fujimoto, J., et al. (1999). Cloning and characterization of DFak56, a homolog of focal adhesion kinase, in Drosophila melanogaster. J. Biol. Chem. 274(41): 29196-201. PubMed Citation: 10506176

Gilmore, A. P. and Romer, L. H. (1996). Inhibition of focal adhesion kinase (FAK) signaling in focal adhesions decreases cell motility and proliferation. Mol. Biol. Cell 7: 1209-1224. Medline abstract: 8856665

Grabbe, C., et al. (2004). Focal adhesion kinase is not required for integrin function or viability in Drosophila. Development 131: 5795-5805. 15525665

Green, H. J., Griffiths, A. G. M., Ylanne, J. and Brown, N. H. (2018). Novel functions for integrin-associated proteins revealed by analysis of myofibril attachment in Drosophila. Elife 7. PubMed ID: 30028294

Gu, J., Tamura, M. and Yamada, K. M. (1998). Tumor suppressor PTEN inhibits integrin- and growth factor-mediated mitogen-activated protein (MAP) kinase signaling pathways. J. Cell Biol. 143(5): 1375-83. PubMed Citation: 9832564

Hackeng, C. M., et al. (1999). Low density lipoprotein phosphorylates the focal adhesion-associated kinase p125(FAK) in human platelets independent of integrin alphaIIb beta3. J. Biol. Chem. 274(1): 384-8. PubMed Citation: 9867854

Han, D. C. and Guan, J. L. (1999). Association of focal adhesion kinase with Grb7 and its role in cell migration. J. Biol. Chem. 274(34): 24425-30.. PubMed Citation: 10446223

Harte, M. T., et al. (1996). p130Cas, a substrate associated with v-Src and v-Crk, localizes to focal adhesions and binds to focal adhesion kinase. J. Biol. Chem. 271(23): 13649-55. PubMed Citation: 8662921

Henry, C. A., et al. (2001). Roles for zebrafish focal adhesion kinase in notochord and somite morphogenesis. Dev. Biol. 240(2): 474-487. 11784077

Hildebrand, J. D., Taylor, J. M. and Parsons, J. T. (1996). An SH3 domain-containing GTPase-activating protein for Rho and Cdc42 associates with focal adhesion kinase. Mol. Cell. Biol. 16: 3169 -3178. Medline abstract: 8649427

Hotchin, N. A., et al. (1999). Differential activation of focal adhesion kinase, Rho and Rac by the ninth and tenth FIII domains of fibronectin. J. Cell Sci. 112 (Pt 17): 2937-46. PubMed Citation: 10444388

Hsia, D. A., Mitra, S. K., Hauck, C. R., Streblow, D. N., Nelson, J. A., Ilic, D., Huang, S., Li, E., Nemerow, G. R., Leng, J. et al. (2003). Differential regulation of cell motility and invasion by FAK. J. Cell Biol. 160: 753-767. 12615911

Huang, Z., et al. (2007). Crk-associated substrate (Cas) signaling protein functions with integrins to specify axon guidance during development. Development 134: 2337-2347. PubMed Citation: 17537798

Hughes, P. E., et al. (1997). Suppression of integrin activation: a novel function of a Ras/Raf-initiated MAP kinase activity. Cell 88: 521-530. PubMed Citation: 9038343

Igishi T., et al. (1999). Divergent signaling pathways link focal adhesion kinase to mitogen-activated protein kinase cascades. Evidence for a role of paxillin in c-Jun NH(2)-terminal kinase activation. J. Biol. Chem. 274(43): 30738-46. PubMed Citation: 10521463

Ilic, D., Furuta, Y., Kanazawa, S., Takeda, N., Sobue, K., Nakatsuji, N., Nomura, S., Fujimoto, J., Okada, M. and Yamamoto, T. (1995). Reduced cell motility and enhanced focal adhesion contact formation in cells from FAK-deficient mice. Nature 377: 539-544. Medline abstract: 7566154

Ilic, D., et al. (1998). Extracellular matrix survival signals transduced by focal adhesion kinase suppress p53-mediated apoptosis. J. Cell Biol. 143(2): 547-60. PubMed Citation: 9786962

Klinghoffer, R. A., et al. (1999). Src family kinases are required for integrin but not PDGFR signal transduction. EMBO J. 18(9): 2459-2471. PubMed Citation: 10228160

Kragtorp, K. A. and Miller, J. R. (2006). Regulation of somitogenesis by Ena/VASP proteins and FAK during Xenopus development. Development 133: 685-695. Medline abstract: 16421193

Kumar, S., et al. (1999). Negative regulation of PYK2/related adhesion focal tyrosine kinase signal transduction by hematopoietic tyrosine phosphatase SHPTP1. J. Biol. Chem. 274(43): 30657-63. PubMed Citation: 10521452

LaLonde, D. P., Grubinger, M., Lamarche-Vane, N. and Turner, C. E. (2006). CdGAP associates with actopaxin to regulate integrin-dependent changes in cell morphology and motility. Curr. Biol. 16: 1375-1385. Medline abstract: 16860736

Lebrun, P., et al. (1998). Insulin receptor substrate-1 as a signaling molecule for focal adhesion kinase pp125(FAK) and pp60(src). J. Biol. Chem. 273(48): 32244-53. PubMed Citation: 9822703

Li, W., et al. (2004). Activation of FAK and Src are receptor-proximal events required for netrin signaling. Nat. Neurosci. 7: 1213-1221. 15494734

Li, X., et al. (1999). Interactions between two cytoskeleton-associated tyrosine kinases: calcium-dependent tyrosine kinase and focal adhesion tyrosine kinase. J. Biol. Chem. 274(13): 8917-24. PubMed Citation: 10085136

Lilja, J., Zacharchenko, T., Georgiadou, M., Jacquemet, G., De Franceschi, N., Peuhu, E., Hamidi, H., Pouwels, J., Martens, V., Nia, F. H., Beifuss, M., Boeckers, T., Kreienkamp, H. J., Barsukov, I. L. and Ivaska, J. (2017). SHANK proteins limit integrin activation by directly interacting with Rap1 and R-Ras. Nat Cell Biol 19(4): 292-305. PubMed ID: 28263956

Liu, G., Beggs, H., Jurgensen, C., Park, H. T., Tang, H., Gorski, J., Jones, K. R., Reichardt, L. F., Wu, J. and Rao, Y. (2004). Netrin requires focal adhesion kinase and Src family kinases for axon outgrowth and attraction. Nat. Neurosci. 7: 1222-1232. 15494732

Liu, Y., Loijens, J. C., Martin, K. H., Karginov, A. V. and Parsons, J. T. (2002). The association of ASAP1, an ADP ribosylation factor-GTPase activating protein, with focal adhesion kinase contributes to the process of focal adhesion assembly. Mol. Biol. Cell 13: 2147-2156. Medline abstract: 12058076

Lyman, S., et al. (1997). Integrin-mediated activation of focal adhesion kinase is independent of focal adhesion formation or integrin activation. Studies with activated and inhibitory beta3 cytoplasmic domain mutants. J. Biol. Chem. 272(36): 22538-22547. PubMed Citation: 9278407

Ma, A., et al. (2001). Serine phosphorylation of Focal adhesion kinase in interphase and mitosis: A possible role in modulating binding to p130Cas. Mol. Biol. Cell 12: 1-12. 11160818

Macagno, J. P., Diaz Vera, J., Yu, Y., Macpherson, I., Sandilands, E., Palmer, R., Norman, J. C., Frame, M. and Vidal, M. (2014). FAK acts as a suppressor of RTK-MAP kinase signalling in Drosophila melanogaster epithelia and human cancer cells. PLoS Genet 10: e1004262. PubMed ID: 24676055

Manes S., et al. (1999). Concerted activity of tyrosine phosphatase SHP-2 and focal adhesion kinase in regulation of cell motility. Mol. Cell. Biol. 19(4): 3125-35. PubMed Citation: 10082579

McLean, G. W., et al. (2005). Specific deletion of focal adhesion kinase suppresses tumor formation and blocks malignant progression. Genes Dev. 18(24): 2998-3003. 15601818

Menegon, A., et al. (1999). FAK+ and PYK2/CAKbeta, two related tyrosine kinases highly expressed in the central nervous system: similarities and differences in the expression pattern. Eur. J. Neurosci. 11(11): 3777-88. PubMed Citation: 10583467

Moissoglu, K. and Gelman, I. H. (2003). v-Src rescues actin-based cytoskeletal architecture and cell motility and induces enhanced anchorage independence during oncogenic transformation of focal adhesion kinase-null fibroblasts. J. Biol. Chem. 278: 47946-47959. 14500722

Murakami, S., et al. (2007). Focal adhesion kinase controls morphogenesis of the Drosophila optic stalk. Development 134: 1539-1548. Medline abstract: 17360775

Nolan, K., Lacoste, J. and Parsons, J. T. (1999). Regulated expression of focal adhesion kinase-related nonkinase, the autonomously expressed C-terminal domain of focal adhesion kinase. Mol. Cell. Biol. 19(9): 6120-9. PubMed Citation: 10454559

Oktay, M., et al. (1999). Integrin-mediated activation of focal adhesion kinase Is required for signaling to Jun NH2-terminal kinase and progression through the G1 phase of the cell cycle. J. Cell Biol. 145: 1461-1470. PubMed Citation: 10385525

Owen, J. D., et al. (1999). Induced focal adhesion kinase (FAK) expression in FAK-null cells enhances cell spreading and migration requiring both auto- and activation loop phosphorylation sites and inhibits adhesion-dependent tyrosine phosphorylation of Pyk2. Mol. Cell. Biol. 19(7): 4806-18. PubMed Citation: 10373530

Padmanabhan, J., Clayton, D. and Shelanski, M. L. (1999). Dibutyryl cyclic AMP-induced process formation in astrocytes is associated with a decrease in tyrosine phosphorylation of focal adhesion kinase and paxillin. J. Neurobiol. 39(3): 407-22. 99290581

Palmer, R. H., et al. (1999). DFak56 is a novel Drosophila melanogaster focal adhesion kinase. J. Biol. Chem. 274(50): 35621-9. PubMed Citation: 10585440

Pandey, P., et al. (1999). Activation of p38 mitogen-activated protein kinase by PYK2/related adhesion focal tyrosine kinase-dependent mechanism. J. Biol. Chem. 274(15): 10140-4. PubMed Citation: 10187797

Polte, T. R. and Hanks, S. K. (1995). Interaction between focal adhesion kinase and Crk-associated tyrosine kinase substrate p130Cas. Proc. Natl. Acad. Sci. 92(23): 10678-82. PubMed Citation: 7479864

Raghavan, S., Vaezi, A. and Fuchs, E. (2003). A role for alphaß1 integrins in focal adhesion function and polarized cytoskeletal dynamics. Dev. Cell 5: 415-427. 12967561

Reiske, H. R., et al. (1999). Requirement of phosphatidylinositol 3-kinase in focal adhesion kinase-promoted cell migration. J. Biol. Chem. 274(18): 12361-6. PubMed Citation: 10212207

Ren, X. R., et al. (2004). Focal adhesion kinase in netrin-1 signaling. Nat. Neurosci. 7: 1204-1212. 15494733

Renshaw, M. W., et al. (1999). Focal adhesion kinase mediates the integrin signaling requirement for growth factor activation of MAP kinase. J. Cell Biol. 147(3): 611-8. PubMed Citation: 10545504

Richardson, A., et al. (1997). Inhibition of cell spreading by expression of the C-terminal domain of focal adhesion kinase (FAK) is rescued by coexpression of Src or catalytically inactive FAK: a role for paxillin tyrosine phosphorylation. Mol. Cell. Biol. 17(12): 6906-6914. PubMed Citation: 9372922

Rodriguez-Fernandez, J. L., et al. (1999). The interaction of activated integrin lymphocyte function-associated antigen 1 with ligand intercellular adhesion molecule 1 induces activation and redistribution of focal adhesion kinase and proline-rich tyrosine kinase 2 in T lymphocytes. Mol. Biol. Cell 10(6): 1891-907. PubMed Citation: 10359604

Ruhl, M., et al. (1999). Soluble collagen VI induces tyrosine phosphorylation of paxillin and focal adhesion kinase and activates the MAP kinase erk2 in fibroblasts. Exp. Cell Res. 250(2): 548-57. PubMed Citation: 10413607

Sagnier, T., Grienenberger, A., Mariol, M., Berenger, H., Pradel, J. and Graba, Y. (2000). Dynamic expression of d-CdGAPr, a novel Drosophila melanogaster gene encoding a GTPase activating protein. Mech. Dev. 94: 267-270. Medline abstract: 10842085

Salazar, E. P. and Rozengurt, E. (1999). Bombesin and platelet-derived growth factor induce association of endogenous focal adhesion kinase with Src in intact Swiss 3T3 cells. J. Biol. Chem. 274(40): 28371-8. PubMed Citation: 10497197

Sastry, S. K., et al. (1999). Quantitative changes in integrin and focal adhesion signaling regulate myoblast cell cycle withdrawal. J. Cell Biol. 144(6): 1295-1309. PubMed Citation: 10087271

Schaller, M. D. and Parsons, J. T. (1995). pp125FAK-dependent tyrosine phosphorylation of paxillin creates a high-affinity binding site for Crk. Mol. Cell. Biol. 15(5): 2635-45. PubMed Citation: 7537852

Schaller, M. D, Hildebrand, J. D, and Parsons, J. T. (1999). Complex formation with focal adhesion kinase: A mechanism to regulate activity and subcellular localization of Src kinases. Mol. Biol. Cell 10(10): 3489-505. PubMed Citation: 10512882

Schlaepfer, D. D. and Hunter, T. (1997). Focal adhesion kinase overexpression enhances ras-dependent integrin signaling to ERK2/mitogen-activated protein kinase through interactions with and activation of c-Src. J. Biol. Chem. 272(20): 13189-13195. PubMed Citation: 9148935

Schlomann, U., et al. (2009). The stimulation of dendrite growth by Sema3A requires integrin engagement and focal adhesion kinase. J. Cell Sci. 122(Pt 12): 2034-42. PubMed Citation: 19454481

Shen, T. L., et al. (2005). Conditional knockout of focal adhesion kinase in endothelial cells reveals its role in angiogenesis and vascular development in late embryogenesis. J Cell Biol. 169(6): 941-52. 15967814

Sorenson, C. M. and Sheibani N. (1999). Focal adhesion kinase, paxillin, and bcl-2: analysis of expression, phosphorylation, and association during morphogenesis. Dev. Dyn. 215(4): 371-82. PubMed Citation: 10417825

Tahiliani, P. D., et al. (1997). The role of conserved amino acid motifs within the integrin beta3 cytoplasmic domain in triggering focal adhesion kinase phosphorylation. J. Biol. Chem. 272(12): 7892-7898. PubMed Citation: 9065456

Tamura, M., et al. (1999). PTEN interactions with focal adhesion kinase and suppression of the extracellular matrix-dependent phosphatidylinositol 3-kinase/Akt cell survival pathway. J. Biol. Chem. 274(29): 20693-703>. PubMed Citation: 10400703

Thomas, J. W., et al. (1999). The role of focal adhesion kinase binding in the regulation of tyrosine phosphorylation of paxillin. J. Biol. Chem. 274(51): 36684-92. PubMed Citation: 10593973

Tikhmyanova, N., et al. (2010). Dcas supports cell polarization and cell-cell adhesion complexes in development. PLoS One 5(8): e12369. PubMed Citation: 20808771

Tsai, P. I., Wang, M., Kao, H. H., Cheng, Y. J., Lin, Y. J., Chen, R. H. and Chien, C. T. (2012). Activity-dependent retrograde laminin A signaling regulates synapse growth at Drosophila neuromuscular junctions. Proc Natl Acad Sci 109(43): 17699-704. PubMed Citation: 23054837

Tsai, P. I., et al. (2008) Fak56 functions downstream of integrin alphaPS3βν and suppresses MAPK activation in neuromuscular junction growth. Neural Dev. 3: 26. PubMed Citation: 18925939

Tsuchida, M., et al. (1999). T cell activation up-regulates the expression of the focal adhesion kinase Pyk2: opposing roles for the activation of protein kinase C and the increase in intracellular Ca2+. J. Immunol. 163(12): 6640-50. PubMed Citation: 10586059

Tsuchida, M., et al. (2000). Regulation of T cell receptor- and CD28-induced tyrosine phosphorylation of the focal adhesion tyrosine kinases pyk2 and fak by protein kinase C. A role for protein tyrosine phosphatases. J. Biol. Chem. 275(2): 1344-50. PubMed Citation: 10625683

Tsutsumi, R., et al. (2006). Focal adhesion kinase is a substrate and downstream effector of SHP-2 complexed with Helicobacter pylori CagA. Mol. Cell. Biol. 26(1): 261-76. 16354697

Turner, C. E. and Miller, J. T. (1994). Primary sequence of paxillin contains putative SH2 and SH3 domain binding motifs and multiple LIM domains: identification of a vinculin and pp125Fak-binding region. J. Cell Sci. 107 ( Pt 6): 1583-91. PubMed Citation: 7525621

van de Water, B., Nagelkerke, J. F. and Stevens, J. L. (1999). Dephosphorylation of focal adhesion kinase (FAK) and loss of focal contacts precede caspase-mediated cleavage of FAK during apoptosis in renal epithelial cells. J. Biol. Chem. 274(19): 13328-37. PubMed Citation: 10224094

Webb, D. J., Donais, K., Whitmore, L. A., Thomas, S. M., Turner, C. E., Parsons, J. T. and Horwitz, A. F. (2004). FAK-Src signalling through paxillin, ERK and MLCK regulates adhesion disassembly. Nat. Cell Biol. 6: 154-161. 14743221

Wen, L. P., et al. (1997). Cleavage of focal adhesion kinase by caspases during apoptosis. J. Biol. Chem. 272(41): 26056-61. PubMed Citation: 9325343

Wu, X., Gan, B., Yoo, Y. and Guan, J. L. (2005). FAK-mediated Src phosphorylation of Endophilin A2 inhibits endocytosis of MT1-MMP and promotes ECM degradation. Dev. Cell 9(2): 185-96. 16054026

Zhai, J., Lin, H., Nie, Z., Wu, J., Canete-Soler, R., Schlaepfer, W. W. and Schlaepfer, D. D. (2003). Direct interaction of focal adhesion kinase with p190RhoGEF. J. Biol. Chem. 278: 24865-24873. Medline abstract: 12702722

Zhang, X., et al. (1999). Focal adhesion kinase promotes phospholipase C-gamma1 activity. Proc. Natl. Acad. Sci. 96(16): 9021-6. PubMed Citation: 10430888

Zhang, Z., et al. (1999). Cytoskeleton-dependent tyrosine phosphorylation of the p130(Cas) family member HEF1 downstream of the G protein-coupled calcitonin receptor. Calcitonin induces the association of HEF1, paxillin, and focal adhesion kinase. J. Biol. Chem. 274(35): 25093-8. PubMed Citation: 10455189

Zhao, J. H, Reiske, H. and Guan, J. L. (1998). Regulation of the cell cycle by focal adhesion kinase. J. Cell Biol. 143(7): 1997-2008. PubMed Citation: 12820964

Zhao, J., Zheng, C. and Guan, J. (2000). Pyk2 and FAK differentially regulate progression of the cell cycle. J. Cell Sci. 113: 3063-3072. 10934044

Zhao, J., et al. (2003). Identification of transcription factor KLF8 as a downstream target of focal adhesion kinase in its regulation of Cyclin D1 and cell cycle progression. Molec. Cell 11: 1503-1515. 12820964

Zheng, C., et al. (1998). Differential regulation of Pyk2 and focal adhesion kinase (FAK). The C-terminal domain of FAK confers response to cell adhesion. J. Biol. Chem. 273(4): 2384-9. PubMed Citation: 9442086


Focal adhesion kinase-like: Biological Overview | Evolutionary Homologs | Regulation | Developmental Biology | Effects of Mutation

date revised: 2 December 2018

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

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