(2014) 33, 4537–4547 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc

REVIEW SRChing for the substrates of Src

AB Reynolds1, SB Kanner2, AH Bouton3, MD Schaller4, SA Weed5, DC Flynn6 and JT Parsons7

By the mid 1980’s, it was clear that the transforming activity of oncogenic Src was linked to the activity of its domain and attention turned to identifying substrates, the putative next level of control in the pathway to transformation. Among the first to recognize the potential of phosphotyrosine-specific antibodies, Parsons and colleagues launched a risky shotgun-based approach that led ultimately to the cDNA and functional characterization of many of today’s best-known Src substrates (for example, p85-Cortactin, p110-AFAP1, p130Cas, p125FAK and p120-catenin). Two decades and over 6000 citations later, the original goals of the project may be seen as secondary to the enormous impact of these substrates in many areas of biology. At the request of the editors, this review is not restricted to the current status of the substrates, but reflects also on the anatomy of the project itself and some of the challenges and decisions encountered along the way.

Oncogene (2014) 33, 4537–4547; doi:10.1038/onc.2013.416; published online 14 October 2013 Keywords: Src; cortactin; p130Cas; FAK; p120-catenin; AFAP1

INTRODUCTION and/or visualized by western blotting. The technology was rapidly The decade of the 1980’s witnessed an immense leap in the adopted by Parsons and colleagues in the form of a risky endeavor understanding of oncogenic signaling, including that mediated that led ultimately to the cDNA cloning and functional by v-Src, the first identified oncoprotein1 and the first described characterization of several of today’s best-known Src substrates. proteintyrosinekinase(PTK).2 The discovery of Src as a PTK was Contrary to expectations, most shared an ability to regulate the quickly followed by identification of other oncogenic PTKs3,4 actin , as exemplified by the roles of FAK and p120 in including members of the family, and cell–ECM and cell–cell adhesion, respectively. They were, in fact, attention turned to the possibility of downstream effectors. The among the first representatives of a larger network of substrates search for receptor tyrosine kinase substrates was facilitated by that now define one of the better-recognized roles of c-Src. the ligand-induced formation of stable receptor–substrate At the request of the editors, this review is not entirely about complexes that could be immunoprecipitated for analysis. In the substrates, but reflects also on the anatomy of the project contrast, the substrates of Src were slow to emerge owing itself and some of the challenges and decisions encountered primarily to technical constraints. Nonetheless, by mid-decade it along the way. Indeed, despite the enormous long-term impact of was clear that the kinase activity of Src was essential for its the work described below, it was not at all obvious at the time that transforming activity and the possibility of a substrate-depen- the spectrum of apparent Src targets, as indicated by pTyr- dent pathway to transformation was increasingly difficult to antibodies, would turn out to be anything more than a collection ignore. of physiologically irrelevant , phosphorylated unproduc- Lacking the handle provided by today’s phosphotyrosine- tively by an overexpressed and promiscuous kinase. Arguing specific antibodies, identifying putative Src substrates was a slow against that notion was the apparent specificity implied by the and labor-intensive process, requiring high-level 32P-labeling of recognition of just 15–20 well-defined pTyr substrates from a Src-transformed cells, separation of the radiolabelled phospho- proteome of some 30 000 candidates. proteins by 2-D polyacrylamide gel electrophoresis and an in-gel alkaline treatment protocol designed to leave phosphotyrosine residues intact, while cleaving phospho-serine and -threonine. It The search for Src substrates was largely ineffective and by 1988, only a handful of putative In 1987, postdoctoral fellows Kanner and Reynolds, together with substrates had been identified (for example, b1-, talin, Parsons, initiated a high-risk project (never subjected to NIH peer vinculin, phosphatidylinositol-3 kinase, p36 (calpactin) and p42 review) to identify the Src substrates indicated by pTyr-Abs. The (later, identified as MAP kinase)).5 two-pronged strategy was to first use pTyr-antibodies to purify the The technical problems were ultimately resolved by the tyrosine-phosphorylated proteins from Src-transformed cell discovery of phosphotyrosine (pTyr)-specific antibodies,6 which lysates, with the aim to shotgun immunize mice for generation provided a convenient and highly specific molecular handle of monoclonal antibodies (mAbs) to individual substrates. The on pTyr-containing proteins. For the first time, tyrosine- substrates were then to be identified directly by cDNA cloning, phosphorylated proteins could be selectively immunoprecipitated using the mAbs to screen lGT11 expression libraries.

1Department of Biology, Vanderbilt University, Nashville, TN, USA; 2Arrowhead Research Corporation, Madison, WI, USA; 3Departments of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA; 4Department of Biochemistry, 3124 HSN, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV, USA; 5Department of Neurobiology and Anatomy, 1833 Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV, USA; 6Department of Medical Lab Sciences, College of Health Sciences, University of Delaware, Newark, DE, USA and 7Departments of Microbiology, Immunology and Cancer Biology, University of Virginia Cancer Center, Charlottesville, VA, USA. Correspondence: Dr DC Flynn, College of Health Sciences, University of Delaware, Newark, DE 19716, USA. E-mail: dfl[email protected] Received 31 May 2013; revised 16 August 2013; accepted 17 August 2013; published online 14 October 2013 Substrates of Src AB Reynolds et al 4538 Accordingly, tyrosine-phosphorylated proteins were purified However, a much better measure of the importance of the from cell lysates of Src-transformed chicken embryo fibroblasts, substrates stems from their enormous impact over the past two essentially in one step, using anti-pTyr immunoaffinity chromato- decades in areas of biology that extend well beyond the early graphy.7 Given the small amount of purified protein available, only focus on Src. four mice were immunized, two of which responded robustly to the chicken-derived antigens. As there was almost no information on substrate abundance, direct and indirect ELISAs were P85/CORTACTIN developed, the former for robustness, the latter providing Early work with p85 revealed striking localization to the cortical increased sensitivity.8 Both mice were fused, generating 786 actin cytoskeleton, giving rise to the name ‘cortactin’.9 On western hybridomas, of which 251 were positive in one or both of the blots, cortactin appeared as two to three bands ranging from ELISA assays. After subcloning and biochemical validation, the 80–85 kDa, all of which were phosphorylated on tyrosine by final tally came to 32 individual mAbs recognizing seven distinct Src. Early work in several cell types revealed that cortactin is tyrosine-phosphorylated substrates, and in a few cases, pTyr also tyrosine-phosphorylated by other Src-family members and itself.8 following ligand activation of the FGF receptor.10 To isolate the corresponding cDNAs efficiently, lGT11 libraries were initially screened with an antibody pool comprised of the Structure best mAbs to each substrate. The positive clones were then Cortactin was described well ahead of the other substrates due rescreened with each individual antibody, resulting in partial 11 cDNA clones for most substrates, the lone exception being p120 largely to the rapid isolation of a full-length cDNA clone. (as described elsewhere). Because of the rapid expansion in the Sequence analysis revealed a novel amino-terminal domain scope of the project, it was broken up according to substrate consisting of six-and-one-half 37 amino-acid tandem repeats, (along with corresponding mAbs and partial cDNAs), and later identified as the actin-binding region, and a carboxy-terminal redistributed to lab members to be completed as independent SH3 domain. The human ortholog (EMS1) was cloned shortly thereafter as a component of the 11q13 amplicon in head and projects. 12 Interestingly, sequencing of the open reading frames revealed neck squamous cell carcinoma (HNSCC). that all but tensin encoded entirely novel . Moreover, with the possible exception of p185, which was not pursued, all of the Cellular function and effects of proteins were ultimately validated as physiologically relevant Src Consistent with a role in cortical actin regulation, cortactin substrates. Notably, an unexpected finding at the time of original promotes actin polymerization through direct binding to the sequencing was the almost ubiquitous presence of one or more actin related protein (Arp)2/3 complex13–15 and interactions with Src homology domains (for example, SH1, SH2 and SH3). For the Arp2/3 nucleation activator protein, neuronal Wiskott–Aldrich example, FAK itself emerged as an unconventional PTK (that is, Syndrome protein (N-WASp).16,17 Cortactin also stabilizes binding SH1 domain), tensin was the first reported example of an SH2 of Arp2/3 to F-actin networks, consistent with its function in domain in a cytoskeletal protein, and both Cortactin and p130CAS lamellipodia dynamics.18–20 Studies conducted with the N-WASp contained SH3 domains. P120, on the other hand, was similar to activating proteins WIP and Nck1 demonstrate that tyrosine b-catenin, as both proteins contained central Armadillo Repeat phosphorylation of cortactin enhances Arp2/3 activation through Domains. P120 was initially viewed as an outlier based on the lack binding of the adaptor protein Nck1, which in turn activates of PTK-associated domains. However, both p120 and b-catenin are N-WASp directly or through the association of the cortactin–Nck1 prominent Src substrates that together control the dynamic complex with WIP.21 Further work on invadopodia formation interface between classical cadherin complexes and the actin confirmed that Nck1 binding to cortactin is critical for promoting cytoskeleton. Importantly, they bind and regulate all 26 members cytoplasmic N-WASp-mediated Arp2/3 actin nucleation.22,23 These of the human classical cadherin family, thereby accounting for observations indicate a positive role for Src-mediated cortactin substantial control over cell–cell adhesion systems in essentially phosphorylation in regulating Arp2/3-based actin filament all solid tissues. Thus, despite distinctly different roles, all of formation. the substrates share close functional connections to the actin Changes in cellular actin filament polymerization are hallmarks cytoskeleton. of Src-transformation and cortactin is well positioned to regulate The substrates are now well known and firmly established in these changes. Invadosomes (aka, podosomes and invadopo- the literature. The following sections contain brief accounts dia),14 comprise another hallmark of cancer found in most of progress to date for each of the five novel substrates (Table 1). carcinoma cell types, where they direct proteolytic degradation As they were discovered as part of a larger effort to understand of the underlying basement membrane, thereby facilitating local the mechanism(s) underlying transformation by Src, each section tumor cell spread.24,25 Cortactin expression and Src-based at least briefly reflects on the nature of that relationship. phosphorylation are essential for invadosome formation and

Table 1. Tyrosine-phosphorylated substrates

Substrate Cellular function Possible role in transformation identified

p85-Cortactin Dynamic regulation in actin filaments Promotes invadosomes formation, stabilizes lamellipodia p110-AFAP1 Actin filament cross-linking Regulation of Src activity, cell contractility and invadosomes stability p130Cas Protein interactions in complexes and at membrane Regulation of cell adhesion and cell survival protrusions p125FAK Regulation of signaling in focal adhesion complexes Regulation of cell adhesion, survival, and cell–cell contacts p120CTN Regulation of cell–cell adhesion by classical cadherins Regulation of cell–cell contacts The five substrates identified during the course of this study, their cellular function and potential role in Src-directed transformation.

Oncogene (2014) 4537 – 4547 & 2014 Macmillan Publishers Limited Substrates of Src AB Reynolds et al 4539 function,22,26,27 suggesting that tyrosine phosphorylation of dynamic changes in actin filament cross-linking is regulated by cortactin is a critical regulator of the early metastatic cascade. PKCa-mediated phosphorylation, which modulates the con- Indeed, cortactin is now a widely accepted marker for formation and multimer size of AFAP1 in a fashion analogous to invadosomes and may have a role in regulating the dynamic of the Lzip motif.44 Thus, AFAP1 mediates dynamic changes in actin polymerization required for invadopodia function changes in actin filament cross-linking in response to PKCa in transformed cells. phosphorylation. Interestingly, several lines of evidence suggest that AFAP1 can Mouse models and human disease function as an upstream activator of Src. For example, deletion of the AFAP1 Lzip motif enables autonomous activation of c-Src in Interestingly, embryo formation was blocked by constitutive cells.46 In a potentially analogous scenario, PKCa can promote Src cortactin knockout (using trap technology) due to mis- activation by an AFAP1-dependent mechanism. Overall, the data localization of the Arp2/3-F-actin network and inability of the 28 suggest that AFAP1 phosphorylation by PKCa induces structural oocyte to undergo proper asymmetric . In adult changes (for example, destabilization of AFAP1 multimers) that mice, however, the effect of global disruption of the gene potentiate both its actin-filament cross-linking activity and its (induced by Cre-lox activation) was relatively mild, the main positive effects on c-Src.47–50 A working model based on these phenotype being increased vascular permeability. A possible data proposes that the Lzip motif functions as an auto inhibitor explanation for the discrepancy is that expression of the amino- of AFAP1 activity by stabilizing the multimeric configuration, terminal end of cortactin by the gene trap vector may have a a condition relieved by upstream activation of PKCa. Notably, dominant-negative affect, as apposed to the null phenotype 29 AFAP1 knockdown has little effect on the Src-transformation associated with the conditional knockout. phenotype. Thus, AFAP1 is apparently an upstream effector of EMS1 amplification occurs in multiple carcinoma types, suggest- 30 c-Src, but its role downstream of activated Src, does not modulate ing a role for cortactin in human malignancy. In HNSCC, increased transformation. copy number correlates with poor patient survival, increased recurrence and lymph node involvement.31,32 In premalignant HNSCC lesions, EMS1 overexpression associates with progression to Mouse models and human cancer malignancy.33 Other effects of ems1 gene amplification in HNSCC To date, AFAP1 has not been extensively investigated with regard include increased invasive disease, Arp2/3 activation, invadopodia- to potential roles in human cancer. Its expression, however, is associated matrix degradation, metalloproteinase secretion and significantly elevated in at least two , namely cancer of the resistance to targeted therapeutics and radiation.34–37 These prostate and neuroblastoma.51,52 Together, these data indicate observations suggest a prominent role for cortactin amplification that AFAP1 may be positioned to promote cancer progression, and/or overexpression in HNSCC. possibly through its function as a signaling protein or as a regulator of cytoskeletal integrity and cell contractility. Its role as a c-Src activating protein and regulator of stress filament cross- P110/AFAP1 linking in tumor formation in mouse models has not been p110/AFAP1 (formerly AFAP-110) was initially identified as a determined. The establishment of mouse models will be 110-kDa tyrosine-phosphorylated protein that bound to activated important to determine a role for AFAP1 in cancer progression. forms of Src in an SH3- and SH2-dependent fashion.7,8,38 Immuno- Early results indicate that AFAP1 knockout mice appear to have fluorescence analysis of normal cells revealed an association with deficits in lactation, indicating AFAP1’s normal function could stress filaments and the cortical actin matrix, giving rise to the have an important role in breast physiology (Cho, Flynn and name ‘AFAP-110’ (actin filament-associated protein 110 kDa).8,38 It Cunnick, manuscript in preparation). This mouse model may also was later re-named ‘AFAP1’ (actin filament-associated protein-1) be a useful tool to determine a role for AFAP1 in tumor formation by the Project to distinguish it from newly or tumor progression. discovered family members AFAP1L1 (AFAP1-like 1) and AFAP1L2 (AFAP1-like 2).39,40 P130CAS (CAS) Of the 15–20 Src substrates evident on the original pTyr western Structure blots, p110 (AFAP1) and p130Cas (Cas) were singled out because cDNA library screening initially led to the discovery of the it strongly coimmunoprecipitated with Src, a potential indicator neuronal form of AFAP1, a 120-kDa protein called AFAP-120 of physiological relevance.8,38,53 The p130 interaction turned out expressed exclusively in neurons.41 Further screening turned up to be dependent on the Src SH2 domain, suggesting an SH2- the fibroblast form of AFAP1, which lacks just one codon relative mediated interaction with a phosphotyrosine epitope on p130. to the neuronal form.42 Structure–function analyses revealed an This same p130 protein also associated with the v-Crk actin filament binding motif in the AFAP1 carboxy-terminus,43,44 oncoprotein (p47v-Crk). The p130 cDNA was later cloned by preceded by a series of protein-binding modules, including 2 Hirai et al.,54 who proposed the name p130Cas for ‘Crk-associated pleckstrin homology domains, one of which binds to the protein substrate’. kinase C (PKC) isoforms a, b, g and l. AFAP1 also contained SH2-binding motifs (generated by Src phosphorylation) and an Structure SH3-binding motif that facilitates stable interactions with Src, as well as the Src-family members Fyn and Lyn.41,45 These data Analysis of the amino-acid sequence of p130Cas (Cas) predicted a indicate that AFAP1 may function by bridging interactions unique domain structure consisting of an amino-terminal SH3 between Src-family kinases and the actin cytoskeleton. domain and a centrally located ‘substrate-binding domain’ containing 15 Tyr-X-X-Pro motifs hypothesized to serve as binding sites for the Crk SH2 domains when phosphorylated. The carboxy- Cellular function and effects of phosphorylation terminus contained an additional predicted Src SH2 domain- In addition to direct interactions with actin filaments, AFAP1 can and an adjacent poly-proline motif likely to function multimerize via a carboxy-terminal leucine motif43,44 to function as an SH3 domain-binding site for Src.55 Analysis of the dynamic as an actin filament cross-linking protein. Deletion of this motif interaction between Src and Cas led to a novel paradigm of reduces multimer size, and induces a transition from cortical fibers processive binding between Src and its substrates, whereby the to tighter actin filament bundles. The ability of AFAP1 to affect kinase forms an initial, lower affinity interaction with the substrate

& 2014 Macmillan Publishers Limited Oncogene (2014) 4537 – 4547 Substrates of Src AB Reynolds et al 4540 (in this case via the Cas SH3 domain) before enzymatic response to antiestrogens and cytotoxic agents.84,89,90 The ability modification of the substrate.56,57 Ensuing phosphorylation of of Cas to bind to, and activate, c-Src62,91,92 has an important role in the adjacent tyrosine residue on Cas then provides a binding site this resistance pathway89,90 demonstrating a clear link between for the Src SH2 domain, significantly strengthening the interaction Cas, Src and cell survival. between the two proteins.55 A similar model was predicted for AFAP1, whereby SH3-directed interactions were required for tyrosine phosphorylation of AFAP1 and high-affinity binding FOCAL ADHESION KINASE, P125FAK (FAK) with Src.58,59 Binding of Cas to the c-Src SH3 domain also results Early studies demonstrated that in normal chick embryo in release of c-Src’s autoinhibitory conformation resulting in fibroblast cells, the only major substrate phosphorylated on activation of the kinase.39,56,60–62 Thus, as with AFAP1, p130Cas tyrosine was a single 125 kDa phosphoprotein.93 Ahintregarding can function as an upstream activator of c-Src. Cas is now the function of p125 came from observations that levels of recognized as the prototype of a four-member family of adaptor/ phosphotyrosine increased in integrin-dependent adhesion scaffolding proteins that share common structural and functional structures upon attachment of cells to fibronectin,94 cross- features.63–66 In addition, to Cas (also known as BCAR1 for breast linking cell surface integrins95 or platelet activation and adhesion cancer anti- resistance-1), this family includes NEDD9 to fibrinogen.96 (also known as HEF1 and Cas-L), EFS (also known as Sin) and CASS4 (also known as HEPL). Structure Identification of the p125 protein elicited excitement during the Cellular function and effects of phosphorylation early phases of this project. At that time, BLAST databases As with several of the other Src substrates discussed in this review, came into use and early identification of coding sequences Cas becomes phosphorylated in response to integrin engagement, revealed that p125 was a tyrosine kinase. Further, studies by localizes to focal adhesions and has a critical role in adhesion- other colleagues in the field demonstrated that integrin dependent cytoskeletal remodeling and cell migration.67–72 These engagement was associated with increased tyrosine phosphor- activities are mediated by the processive phosphorylation of ylation in focal adhesion complexes. With p125 thought to have a tyrosine residues located in the Cas substrate-binding domain and role in regulating integrin engagement and signaling, the lab subsequent association with Crk, leading to activation of Rac1 gave much thought into naming the protein, ultimately deciding and Rap1.73–75 Cas-Crk coupling may also contribute to its function on focal adhesion kinase (FAK), due to its colocalization with as a mechanotransducer, as Cas phosphorylation is signi- focal adhesion complexes and intrinsic kinase activity. The ficantly enhanced under conditions in which the Cas substrate- observation that FAK localized to regions of the cell that closely binding domain is physically stretched in response to integrin adhere to the indicated that it might function engagement.76 Cas signaling is involved in the regulation of cell in the transduction of biochemical signals both in response to death once apoptosis has been triggered, which is mediated by a and in the control of cell adhesion.97 The domain organization of 31-kDa carboxy-terminal Cas fragment produced by the action of FAK consists of a FERM (Band Four.one/Ezrin/Radixin/Moesin) apoptosis-induced proteases.77,78 Independent expression of the domain, catalytic domain, a domain of unknown structure 31 kDa fragment causes diverse pro-apoptotic cellular activities. containing several proline-rich regions and a C-terminal In addition to adhesion signaling, Cas has been implicated in domain that mediates localization (the focal adhesion targeting the control of cell proliferation and survival. Cas tyrosine domain).98 phosphorylation is induced in response to a number of 79,80 mitogenic stimuli. However, the mechanisms by which Cas Cellular function and effects of phosphorylation phosphorylation regulates cell proliferation are complex and not Tyrosine 397 is a major site of phosphorylation in FAK and has a yet clear. Cas becomes dephosphorylated on tyrosine and critical role in signaling as a docking site for SH2 domain- phosphorylated on serine/threonine residues during , containing proteins, including Src.97,99 The relationship between which leads to disruption of Cas/FAK/c-Src interactions and FAK and cell adhesion is complex. Integrin-dependent cell dissolution of focal adhesions.81 These changes in adhesion to extracellular matrix proteins stimulates FAK activity phosphorylation may enable Cas to affect dynamic changes in resulting in tyrosine phosphorylation on multiple residues.98 FAK focal adhesions in response to phosphorylation by Src and controls a number of biological responses regulated by cell subsequent dephosphorylation. adhesion, including anoikis, proliferation and cell migration. One role of FAK in the control of cell migration is the regulation of the Mouse models and human disease assembly and disassembly of focal adhesions, adhesive structures Mouse models of breast tumorigenesis further support a role for that tether cells to the underlying extracellular matrix.100–102 FAK Cas in cell proliferation and survival, as mammary gland-specific mutants that cannot be phosphorylated at tyrosine 397 are overexpression of Cas results in tissue hyperplasia coincident defective for disassembly of cell/matrix adhesions. In this regard, with Src and Akt activation.82 Furthermore, when crossed with a Src is likely an important signaling partner for FAK, as Src null cells HER2-neu mammary tumor model, Cas overexpression results in exhibit a similar phenotype.102 shortened tumor latency, while knockdown of Cas in cells isolated FAK regulates cell adhesion through multiple mechanisms. from HER2-neu tumors results in increased apoptosis.82 As an The FAK/Src complex controls phosphorylation and the activity integral component of signaling pathways that control cyto- of PIP5KCI, an isoform of phosphatidylinositol 50 kinase that skeletal dynamics, adhesion, migration, proliferation, survival and discretely localizes to cell/extracellular matrix adhesions, regulates apoptosis, Cas expression positively correlated with poor the generation of PI(4,5)P2 at these sites, and the subsequent prognoses in a number of human cancers, including breast, assembly of these structures.103 FAK can also regulate cell prostate, ovarian and lung.65,83–88 It remains undetermined adhesion through Rho family GTPases.104–108 The focal adhesion whether these pathological states are a direct consequence of targeting domain of FAK mediates binding to two focal adhesion- Cas function and, if so, whether specific Cas signaling nodes can associated proteins, paxillin and talin, and each has been be targeted as a means of disease management. Nonetheless, Cas implicated in directing FAK localization to focal adhesions and in has clearly shown an ability to enhance cell proliferation and the regulation of cell adhesion.109,110 Paxillin had been identified survival. For example, overexpression of Cas in breast cancer cells as a Src substrate by Glenney et al.111 FAK can directly and can result in a failure to undergo arrest and apoptosis in indirectly (via Src) promote paxillin phosphorylation on tyrosine.

Oncogene (2014) 4537 – 4547 & 2014 Macmillan Publishers Limited Substrates of Src AB Reynolds et al 4541 Like FAK, paxillin is required for proper turnover of cell/matrix P120-CATENIN, P120CTN, P120 adhesions and mutants of paxillin that are defective for tyrosine Interest in p120 was initially based on the profiling of tyrosine- phosphorylation are defective for regulation of adhesion phosphorylated proteins in cells expressing activated forms of Src 102 turnover. The interaction of FAK and talin is also important. that were either transformation-competent or transformation- FAK is required for the recruitment of talin to nascent focal defective. Remarkably, p120 was unique among the substrates in 112 adhesions but not to mature focal adhesions; Talin has a role in that its phosphorylation correlated with cell transformation, recruiting or stabilizing FAK in mature adhesions. The interaction suggesting a possible role in the mechanism of transformation.53 between FAK and talin113 could offer another mechanism for regulating adhesion. Further, FAK may regulate the cleavage of Structure talin by calpain, which is an important mechanism for regulation of adhesion.112,114 p85-Cortactin also binds via its SH3 domain to Most of the substrates emerged as unique regulators of the actin two proline-rich sequences in the C-terminal domain of FAK. cytoskeleton, each possessing one or more domains typically of these sites in FAK impairs its ability to regulate cell/ associated with PTK signaling. In contrast, p120 contained tandem 115 armadillo repeats with weak homology to the Armadillo repeats in matrix adhesion turnover. Although these sequences mediate 139,140 interaction with multiple proteins, cortactin may be the critical b-catenin/Armadillo. Further analysis revealed sets of binding partner for controlling adhesion remodeling.115 FAK also tandem repeats in p120 and several other proteins, leading to regulates recruitment of dynamin to adhesive structures, via a the recognition and naming of the Armadillo Repeat Domain, a direct interaction with the FAK FERM domain or an indirect structure that has emerged over the last decade as an important interaction via a phosphorylated tyrosine residue in the protein–protein interaction scaffold. Notably, the similarity to b- C-terminus of FAK (tyrosine 925), and dynamin has been catenin led to testing for an analogous interaction with classical implicated in the disassembly of adhesions.116,117 Thus, FAK may cadherins. It turned out that p120 and b-catenin interact simultaneously with adjacent motifs in the cadherin cytoplasmic regulate cell adhesion through changes in phosphorylation that 141 affect interactions with different binding partners, including Src. tail. Because of these molecular interactions, p120 was originally called ‘Cas’ for cadherin-associated (Src) substrate.142 FAK has an important role in the organization of the 143 extracellular matrix in tissue culture and the basement membrane That name was soon rescinded in favor of p120-catenin (p120) 118,119 to avoid being confused with the previously described Src in vivo. By regulating expression and secretion of matrix 54 metalloproteinases, FAK also controls remodeling of the matrix substrate p130Cas. after its initial organization, which enables FAK to control cell invasion.120,121 FAK functions in regulating the assembly of Cellular function and effects of phosphorylation adhesive structures that mediate matrix degradation. FAK The catenin identifier turned out to be a highly accurate interactions with Cas are required to regulate invadosome descriptor, as p120 and b-catenin are direct stoichiometric binding formation in several cell types, and the subsequent degradation partners for each of the 26 classical cadherins, important of the underlying extracellular matrix.122 FAK also functions in mediators of cell–cell adhesion in solid tissues. b-catenin interacts controlling extracellular matrix degradation at focal adhesions and further with a-catenin, a vinculin-like actin-binding protein at the Cas is an important FAK-binding partner in controlling matrix dynamic functional interface between the cadherin cytoplasmic degradation at these sites as well.123 FAK is implicated in tail and the underlying actin cytoskeleton. The term catenin (that controlling cell proliferation and degradation of the extracellular is, chain or to link), coined well before the discovery of p120, was matrix in response to mechanical signals generated by stiff aimed originally at the a-catenin/b-catenin partnership. However, matrices.124–126 p120 also modulates the actin interface, albeit via direct and/or Although FAK’s role in controlling cell/extracellular matrix indirect regulation of Rho GTPases. For example, cytoplasmic p120 adhesion was not surprising, the finding that FAK has a role in functions as a guanine nucleotide dissociation inhibitor, binding controlling cell–cell adhesions was unexpected. FAK is required for RhoA and blocking its activity by preventing GTP/GDP efficient assembly of N-cadherin- and E-cadherin-mediated cell/ exchange.144,145 Cadherin-bound p120, on the other hand, does cell adhesions and FAK-dependent control of the activity of Rho not bind RhoA directly,144 but instead recruits factors such as family proteins appears responsible for modulating the assembly p190RhoGAP146–148 and ROCK149 that in turn localize RhoA activity of cell/cell junctions.127,128 Impairment of FAK activity also retards to adherens junctions. Hence, p120 and b-catenin are important the disassembly of cell/cell junctions in response to stimuli that regulators of the actin cytoskeleton and participate actively in the promote epithelial to mesenchymal transition.129 Thus, FAK is global integration of cytoskeletal activity. implicated in controlling both the assembly and disassembly of More recently, p120 has emerged as a master regulator of different classes of cell adhesions. classical cadherin stability and cell surface retention. p120 modulates the strength of cell–cell adhesion, in part, by controlling cadherin levels. Nascent cadherin complexes Mouse models and human disease arrive at the cell surface independent of p120. Thereafter, direct FAK is essential for mouse development, as demonstrated by the interaction with p120 is essential as the complex is otherwise embryonic lethal phenotype of FAK-null mice.130 These embryos rapidly internalized and degraded. Thus, loss of p120 in most exhibit defects in mesoderm development, neural tube closure adhesive cell types results in reduced or complete loss of and heart tube formation.131 The analysis of conditional knockout cell–cell adhesion. Mechanistically, p120 prevents cadherin mice has provided additional insight into FAK’s role in endocytosis by physically masking crucial endocytic signals in development. These include essential roles in embryonic the cadherin juxtamembrane domain.150 Recent evidence angiogenesis,132,133 development of the heart/aorta134 and suggests a second role, however, as the cell fate and endocytic differentiation of cells.135 Further, in mouse models adaptor protein Numb can regulate endocytosis of the entire FAK has also been implicated in the regulation of axonal cadherin complex (p120 included) through direct interaction branching,136 myelination137 and mammary gland development with p120.151 during pregnancy.138 As outlined below, FAK is implicated in a Transformation by Src in many cell types is associated with number of human diseases. Thus, FAK appears to have an disruption of adherens junctions, actin-based cell–cell adhesion important role in regulating cell adhesion, cell–cell interactions complexes organized by classical cadherins. p120 is also tyrosine- and cell survival, all of which are altered in Src-transformed cells phosphorylated by various growth factor receptors, including the and in human cancer. EGF, PDGF and CSF-1 receptors,152,153 which are functionally

& 2014 Macmillan Publishers Limited Oncogene (2014) 4537 – 4547 Substrates of Src AB Reynolds et al 4542 coupled to cadherins by poorly understood mechanisms.154 competitive, including , and saracatinib, while The role of p120 phosphorylation in these processes is not yet KX-01 is peptidomimetic.169 Additional non-selective kinase known. p120 itself, on the other hand, is clearly essential for inhibitors have been developed, including (Tasigna), transformation by Src and Rac1, but not H-Ras, it remains an open that inhibit Src-family kinases amongst their off-target activities.170 question, therefore, as to whether tyrosine phosphorylation of Although anti-Bcr-Abl activity has enabled dasatinib, bosutinib and p120 in fact has an essential role in transformation. nilotinib to be marketed for CML, the activity of the four Src inhibitors in clinical trials of solid tumors has been less robust.165,166 Src regulates tumor cell migration, invasion and Mouse models and human disease adhesion in a variety of model systems, suggesting a role in The in vitro effects of p120 loss are also observed in vivo in mouse tumor cell metastasis.171,172 In addition, Src exhibits downstream models, although the complexity of the phenotypes varies activity in both angiogenesis and bone resorption pathways, depending on the epithelial organ targeted. Global p120 knockout indicating functions in tumor cell maintenance and survival.166.Src B is early embryonic lethal ( day 9.5). Targeted deletion in the also has roles in tumor cell proliferation and cell cycle control in intestinal epithelium causes cell–cell adhesion defects and serious both in vitro and in vivo pre-clinical models of many major solid intestinal bleeding, leading to inflammation and death in the first 155 tumor types, including prostate, breast, colon, lung, pancreatic three weeks of life. Curiously, p120 ablation on a very limited and , among others.173,174 The pre-clinical antitumor B basis (that is, 10% of the intestinal epithelial surface) by itself activities of Src inhibitors provided the rationale, which prompted induces intestinal tumors in about half of the mice, but tumors 156 investigators to evaluate Src inhibitors in multiple solid tumor invariably retain at least one p120 allele. In the squamous clinical trials.166 Early trials show promise, leading to advanced epithelia of skin and esophagus, p120 induces an apparently cell studies evaluating combinations with chemotherapy or other autonomous inflammatory response (that is, it cannot be 157,158 targeted agents. One possibility for the limited success of Src attributed to a barrier defect). These clues suggest an as inhibitors as single agents is their inability to induce apoptosis, yet poorly understood role for p120 in the innate immune system. potentially required for clinical antitumor activity, despite their The esophageal phenotype involves progression to esophageal inhibitory effects on tumor cell proliferation and xenograft cancer that closely resembles the human condition. In contrast, growth.173 The significant effects of Src inhibitors in tumor cell the mammary gland does not form at all in the absence of 159 motility in vitro may impart a critical activity on patient metastases. p120, and the salivary gland displays misshapen ducts that lack Previous and ongoing clinical trials evaluating the effects of acini.160 Remarkably, the epithelium of the prostate is not 159 Src inhibitors in metastatic cancer are critical for shaping our noticeably affected, and is the only system to date lacking an understanding of their antitumor capacity. Evaluating early stages obvious phenotype. Despite significant tissue-specific variation, of malignancy such as in adjuvant settings and studying rational the mouse models in general illustrate multiple connections to combinations of Src inhibitors with chemotherapy or other cancer and will ultimately be essential for understanding the roles targeted agents is warranted. Another limitation for the success of p120 misexpression in human cancer. Indeed, the human of Src inhibitors in solid tumors may be a lack of standard pathology literature reveals frequent mislocalization and/or 161 biomarkers. Such data could identify responsive patients, and downregulation of p120 in most epithelial cancers. The clinical trials could focus on those patients that demonstrate a mechanism(s) of p120 dysregulation in human cancer are not molecular response to treatment. The potential for establishing yet clear but the involvement of receptor- and Src-family tyrosine novel Src biomarkers may exist within the rich panel of Src kinases remain a strong possibility. substrates.8 The discovery of Src substrates provided an important cornerstone in the development of agents targeting downstream PHARMACEUTICAL TARGETING OF SRC AND ITS SUBSTRATES effectors of Src function.5 One key target is FAK, implicated in Among the substrates identified in this study, FAK, as well as Src, multiple cancer types.175–178 Similar to its upstream activator Src, appear to have the greatest promise as druggable targets for the FAK promotes tumor cell motility, survival and angiogenesis, treatment of cancer. The approach to identify new signaling thereby validating its potential utility in the clinical setting.175–180 pathway components remains an important element in the Five small-molecule FAK inhibitors have been described, several discovery of targets for pharmaceutical development. However, having advanced into clinical trials.181–183 Although early in the lengthy path from target identification to drug registration development, these agents show promise for use in the shifted the effort toward later stages of preclinical development. armamentarium against different tumor types. Similarly as Companies save time and money if their starting point involves a observed for Src inhibitors, perhaps the pre-clinical validation of preclinically, well-validated target before initiating a drug hunting FAK in survival and cell motility points to the potential activity of campaign.162–164 For these reasons, drug targeting of Src and its inhibitors as potent anti-metastatic agents rather than impacting substrates proves a critical opportunity for the discovery and primary tumors.179–183 Combination therapies that address tumor development of new therapies for cancer and other diseases. de-bulking (for example, chemotherapy) with an anti-metastatic There have been significant strides in developing novel therapies drug (for example, potentially FAK inhibitors) may have the best inhibiting Src.165,166 However, the pharmaceutical industry should patient response rate and progression-free survival. Future and explore new avenues of target discovery to ensure future ongoing clinical evaluation of existing and new inhibitors should development of highly effective treatments. Targeting Src help to answer these fundamental questions. substrates represents a rational next step toward the deliberate Other substrates of Src have been studied exten- intervention of this key signaling cascade in multiple disease sively.5,46,65,68,184–187 The substrate proteins p130Cas, p120Ctn, states. p110-AFAP1, p85-cortactin and p210 tensin exhibit functional To date, four small-molecule inhibitors of Src have been interactions with important partner proteins.5,46,65,68,184–187 Such developed for treatment of various malignancies, including chronic interactions are crucial for their functions in tumor cell biology. myelogenous (CML).165,166 Among these, dasatinib Pharmaceutical intervention of protein–protein interactions (Sprycel) and bosutinib (Bosulif) exhibit low nanomolar activity emerged as a potential area for further exploration.188,189 against Src-family kinases.167,168 However, they both equally inhibit Although many biological therapies are marketed based on Bcr-Abl kinase, the pivotal target in CML, and other tyrosine- disruption or stabilization of protein–protein interactions, the and serine/threonine-specific kinases, thereby promoting their approach remains challenging for small-molecule development anti-CML activity.167–169 Three of the inhibitors are ATP- targeting intracellular proteins. The large surface area between

Oncogene (2014) 4537 – 4547 & 2014 Macmillan Publishers Limited Substrates of Src AB Reynolds et al 4543

Figure 1. The five substrates identified during the course of this study largely regulated cytoskeletal processes that are affected by Src- transformation. P120CTN regulates E-cadherin interactions between cells, p85-cortactin regulates invadopodia formation and actin polymerization, p110-AFAP1 regulates actin filament cross-linking, p125FAK and p130Cas regulate cell adhesion complexes.

protein domains and few binding pockets amenable to small- application of appropriate combinations of new medicines molecule docking limits this approach. scaffolds provided customized for each patient. the basis for generating some protein–protein interface inhibi- tors.188,189 Recently, protein–complex stabilizers that enhance the retention time of key complexes have been generated and may SUMMARY 188,189 have therapeutic utility. Allosteric site modulators offer an Twenty years of study has provided much information about the alternative approach when targeting outside of enzymatic structure and function of Src substrates. The work highlighted domains, such as those that inhibit myristate binding in the herein was among the first to describe an important group of Src 190 autoinhibitory pocket of the Bcr-Abl kinase. A variety of substrates involved in the regulation of cell adhesion and actin potential therapeutic agents have been generated for protein dynamics (Figure 1). Notably, the main roadblock from outset had interfaces that possess high affinity, but are larger than typical little to do with the technical challenges. Indeed, by 1989, 188,189 pharmaceuticals. Overall, the approach has significant merit, just 2 years into the project, both mAbs and partial cDNAs for and the industry is poised to harvest novel medicines from the most of the substrates were already in hand, but it was not a large breadth of protein-complexes involved in tumor cell biology. project for the risk averse. Although the jury is perhaps still out on The Src substrates and their cognate interactions may serve p120, the substrates identified did not turn out to be essential for as targets for pharmaceutical development, possibly by RNA transformation. They were, however, among the first bona fide Src interference, and as clinical biomarkers for drugs that interfere substrates identified, and taught us a great deal about Src and its with Src-associated pathways. substrates. Two decades later, the original goals of the project Targeting Src, FAK or other substrates may provide critical new seem rather unimportant compared to the enormous impact of drugs for cancer treatment, but may best be served in many of the substrates in areas that could not have been combinations with other drugs in parallel or convergent path- anticipated a priori. ways. The development of resistance is common among Driven by a compelling question, application of novel targeted agents, particularly with kinase inhibitors. Some muta- technologies, and the willingness to invest time and energy in a tions in kinases remain sensitive to drugs, while others are ‘risky’ venture, we began the search for the Src substrates. In resistant, such as the T315I mutation of Bcr-Abl not inhibited today’s difficult funding environment, we must consider whether 169 by either dasatinib or bosutinib. Drug combinations that this type of scientific effort will garner support. The substantial include multiple Src pathway inhibitors may provide a rational effort to identify and characterize Src substrates involved not only approach for reducing relapse for malignancies where Src exhibits the authors of this review, but a number of post-doctoral fellows, a direct role. graduate students and technicians. Importantly, the rewards have The discovery of the Src substrates exemplifies a ‘shotgun’ been notable. A survey of PubMed citations lists over 6000 papers 8 approach to identify multiple proteins involved in Src biology. on these substrates discussed above. Understanding the function Today, microarrays, protein arrays, yeast two-hybrid systems, of these proteins far exceeded the original goal of understanding synthetic lethality screens and other technologies enable how Src functions to transform cells and has in addition provided researchers to identify putative targets in multiple settings, novel insights as to how these proteins contribute to the complex including pathways, protein networks and regulation of cellular dynamics. patient responses in clinical trials to experimental agents. Broad approaches to the discovery of critical connections between cellular proteins require exploration to enhance our ACKNOWLEDGEMENTS understanding of the cell system beyond an individual Many thanks to Steven Bernhardt for critical review and suggestions for organization 191 molecular pathway. Defining the system biology and of this review. Also, we are grateful for the help of Jenny Michelle Reed in the understanding tumor microenvironments may hasten the clinical preparation of this review.

& 2014 Macmillan Publishers Limited Oncogene (2014) 4537 – 4547 Substrates of Src AB Reynolds et al 4544 REFERENCES 28 Yu D, Zhang H, Blanpied TA, Smith E, Zhan X. Cortactin is implicated in murine 1 Vogt PK. Retroviral : a historical primer. Nat Rev Cancer. 2012; 12: zygotic development. Exp Cell Res 2010; 316: 848–858. 639–648. 29 Schnoor M, Lai FP, Zarbock A, Klaver R, Polaschegg C, Schulte D et al. Cortactin 2 Hunter T, Sefton BM. Transforming gene product of Rous sarcoma virus phos- deficiency is associated with reduced neutrophil recruitment but increased phorylates tyrosine. Proc Natl Acad Sci USA. 1980; 77: 1311–1315. vascular permeability in vivo. J Exp Med 2011; 208: 1721–1735. 3 Pawson T, Kung TH, Martin GS. Structure and phosphorylation of the Fujinami 30 Wilkerson PM, Reis-Filho JS. The 11q13-q14 amplicon: clinicopathological cor- sarcoma virus gene product. J Virol 1981; 40: 665–672. relations and potential drivers. Genes Cancer 2013; 52: 333–355. 4 Witte ON, Dasgupta A, Baltimore D. Abelson murine leukaemia virus protein is 31 Rodrigo JP, Garcia LA, Ramos S, Lazo PS, Suarez C. EMS1 gene amplification phosphorylated in vitro to form phosphotyrosine. Nature 1980; 283: 826–831. correlates with poor prognosis in squamous cell carcinomas of the head and 5 Schaller MD, Bouton AH, Flynn DC, Parsons JT. Identification and characterization neck. Clin Cancer Res 2000; 6: 3177–3182. of novel substrates for protein tyrosine kinases. Prog Nucleic Acid Res Mol Biol 32 Rodrigo JP, Garcia-Carracedo D, Garcia LA, Menendez S, Allonca E, Gonzalez MV 1993; 44: 205–227. et al. Distinctive clinicopathological associations of amplification of the cortactin 6 Kamps MP, Sefton BM. Identification of multiple novel polypeptide substrates of gene at 11q13 in head and neck squamous cell carcinomas. J Pathol 2009; 217: the v-src, v-yes, v-fps, v-ros, and v-erb-B oncogenic tyrosine protein kinases 516–523. utilizing antisera against phosphotyrosine. Oncogene 1988; 2: 305–315. 33 Rodrigo JP, Alvarez-Alija G, Menendez ST, Mancebo G, Allonca E, Garcia-Carra- 7 Kanner SB, Reynolds AB, Parsons JT. Immunoaffinity purification of tyrosine- cedo D et al. Cortactin and focal adhesion kinase as predictors of cancer risk phosphorylated cellular proteins. J Immunol Methods 1989; 120: 115–124. in patients with laryngeal premalignancy. Cancer Prevention Res 2011; 4: 8 Kanner SB, Reynolds AB, Vines RR, Parsons JT. Monoclonal antibodies to indivi- 1333–1341. dual tyrosine-phosphorylated protein substrates of oncogene-encoded tyrosine 34 Rothschild BL, Shim AH, Ammer AG, Kelley LC, Irby KB, Head JA et al. Cortactin kinases. Proc Natl Acad Sci USA. 1990; 87: 3328–3332. overexpression regulates actin-related protein 2/3 complex activity, motility, and 9 Wu H, Parsons JT. Cortactin, an 80/85-kilodalton pp60src substrate, is a fila- invasion in carcinomas with 11q13 amplification. Cancer Res 2006; mentous actin-binding protein enriched in the cell cortex. J Cell Biol 1993; 120: 66: 8017–8025. 1417–1426. 35 Clark ES, Whigham AS, Yarbrough WG, Weaver AM. Cortactin is an essential 10 Zhan X, Hu X, Friesel R, Maciag T. Long term growth factor exposure and dif- regulator of secretion and extracellular matrix degra- ferential tyrosine phosphorylation are required for DNA synthesis in BALB/c 3T3 dation in invadopodia. Cancer Res 2007; 67: 4227–4235. cells. J Biol Chem 1993; 268: 9611–9620. 36 Timpson P, Wilson AS, Lehrbach GM, Sutherland RL, Musgrove EA, Daly RJ. 11 Wu H, Reynolds AB, Kanner SB, Vines RR, Parsons JT. Identification and char- Aberrant expression of cortactin in head and neck squamous cell carcinoma cells acterization of a novel cytoskeleton-associated pp60src substrate. Mol Cell Biol is associated with enhanced cell proliferation and resistance to the epidermal 1991; 11: 5113–5124. inhibitor gefitinib. Cancer Res 2007; 67: 9304–9314. 12 Schuuring E, Verhoeven E, Mooi WJ, Michalides RJ. Identification and cloning 37 Eke I, Deuse Y, Hehlgans S, Gurtner K, Krause M, Baumann M et al. beta(1)In- tegrin/FAK/cortactin signaling is essential for human head and neck cancer of two overexpressed genes, U21B31/PRAD1 and EMS1, within the resistance to radiotherapy. J Clin Invest 2012; 122: 1529–1540. amplified chromosome 11q13 region in human carcinomas. Oncogene 1992; 7: 38 Kanner SB, Reynolds AB, Wang HC, Vines RR, Parsons JT. The SH2 and SH3 355–361. domains of pp60src direct stable association with tyrosine phosphorylated 13 Uruno T, Liu J, Zhang P, Fan Y, Egile C, Li R et al. Activation of Arp2/3 complex- proteins p130 and p110. EMBO J. 1991; 10: 1689–1698. mediated actin polymerization by cortactin. Nat Cell Biol 2001; 3: 259–266. 39 Xu W, Harrison SC, Eck MJ. Three-dimensional structure of the tyrosine kinase 14 Weaver AM, Heuser JE, Karginov AV, Lee WL, Parsons JT, Cooper JA. Interaction c-Src. Nature 1997; 385: 595–602. of cortactin and N-WASp with Arp2/3 complex. Curr Biol 2002; 12: 1270–1278. 40 Snyder BN, Cho Y, Qian Y, Coad JE, Flynn DC, Cunnick JM. AFAP1L1 is a novel 15 Weed SA, Karginov AV, Schafer DA, Weaver AM, Kinley AW, Cooper JA et al. adaptor protein of the AFAP family that interacts with cortactin and localizes to Cortactin localization to sites of actin assembly in lamellipodia requires inter- invadosomes. Eur J Cell Biol 2011; 90: 376–389. actions with F-actin and the Arp2/3 complex. J Cell Biol 2000; 151:29–40. 41 Flynn DC, Koay TC, Humphries CG, Guappone AC. AFAP-120. A variant form of 16 Kowalski JR, Egile C, Gil S, Snapper SB, Li R, Thomas SM. Cortactin regulates cell the Src SH2/SH3-binding partner AFAP-110 is detected in brain and contains a migration through activation of N-WASP. J Cell Sci 2005; 118(Pt 1): 79–87. novel internal sequence which binds to a 67-kDa protein. J Biol Chem 1995; 270: 17 Martinez-Quiles N, Ho HY, Kirschner MW, Ramesh N, Geha RS. Erk/Src phos- 3894–3899. phorylation of cortactin acts as a switch on-switch off mechanism that controls 42 Flynn DC, Leu TH, Reynolds AB, Parsons JT. Identification and sequence analysis its ability to activate N-WASP. Mol Cell Biol 2004; 24: 5269–5280. of cDNAs encoding a 110-kilodalton actin filament-associated pp60src substrate. 18 Weaver AM, Karginov AV, Kinley AW, Weed SA, Li Y, Parsons JT et al. Cortactin Mol Cell Biol 1993; 13: 7892–7900. promotes and stabilizes Arp2/3-induced actin filament network formation. Curr 43 Qian Y, Baisden JM, Zot HG, Van Winkle WB, Flynn DC. The carboxy terminus of Biol 2001; 11: 370–374. AFAP-110 modulates direct interactions with actin filaments and regulates its 19 Bryce NS, Clark ES, Leysath JL, Currie JD, Webb DJ, Weaver AM. Cortactin pro- ability to alter actin filament integrity and induce lamellipodia formation. Exp Cell motes cell motility by enhancing lamellipodial persistence. Curr Biol 2005; 15: Res 2000; 255: 102–113. 1276–1285. 44 Qian Y, Gatesman AS, Baisden JM, Zot HG, Cherezova L, Qazi I et al. Analysis of 20 Boguslavsky S, Grosheva I, Landau E, Shtutman M, Cohen M, Arnold K et al. p120 the role of the leucine zipper motif in regulating the ability of AFAP-110 to alter catenin regulates lamellipodial dynamics and cell adhesion in cooperation with actin filament integrity. J Cell Biochem 2004; 91: 602–620. cortactin. Proc Natl Acad Sci USA. 2007; 104: 10882–10887. 45 Summy JM, Guappone AC, Sudol M, Flynn DC. The SH3 and SH2 domains are 21 Tehrani S, Tomasevic N, Weed S, Sakowicz R, Cooper JA. Src phosphorylation of capable of directing specificity in protein interactions between the non-receptor cortactin enhances actin assembly. Proc Natl Acad Sci USA. 2007; 104: tyrosine kinases cSrc and cYes. Oncogene 2000; 19: 155–160. 11933–11938. 46 Baisden JM, Qian Y, Zot HM, Flynn DC. The actin filament-associated protein 22 Oser M, Yamaguchi H, Mader CC, Bravo-Cordero JJ, Arias M, Chen X et al. AFAP-110 is an adaptor protein that modulates changes in actin filament Cortactin regulates cofilin and N-WASp activities to control the stages of integrity. Oncogene 2001; 20: 6435–6447. invadopodium assembly and maturation. J Cell Biol 2009; 186: 571–587. 47 Bruce-Staskal PJ, Bouton AH. PKC-dependent activation of FAK and src induces 23 Oser M, Mader CC, Gil-Henn H, Magalhaes M, Bravo-Cordero JJ, Koleske AJ et al. tyrosine phosphorylation of Cas and formation of Cas-Crk complexes. Exp Cell Specific tyrosine phosphorylation sites on cortactin regulate Nck1-dependent Res 2001; 264: 296–306. actin polymerization in invadopodia. J Cell Sci 2010; 123(Pt 21): 3662–3673. 48 Brandt D, Gimona M, Hillmann M, Haller H, Mischak H. C induces 24 Murphy DA, Courtneidge SA. The ‘ins’ and ‘outs’ of podosomes and invadopodia: actin reorganization via a Src- and Rho-dependent pathway. J Biol Chem 2002; characteristics, formation and function. Nat Rev Mol Cell Biol 2011; 12: 413–426. 277: 20903–20910. 25 Linder S, Wiesner C, Himmel M. Degrading devices: invadosomes in proteolytic 49 Gatesman A, Walker VG, Baisden JM, Weed SA, Flynn DC. Protein kinase Calpha cell invasion. Ann Rev Cell Dev Biol 2011; 27: 185–211. activates c-Src and induces podosome formation via AFAP-110. Mol Cell Biol 26 Artym VV, Zhang Y, Seillier-Moiseiwitsch F, Yamada KM, Mueller SC. Dynamic 2004; 24: 7578–7597. interactions of cortactin and membrane type 1 matrix metalloproteinase at 50 Walker VG, Ammer A, Cao Z, Clump AC, Jiang B-H, Kelley LC et al. PI3K activation invadopodia: defining the stages of invadopodia formation and function. Cancer is required for PMA-directed activation of cSrc by AFAP-110. Am J Physiol Cell Res 2006; 66: 3034–3043. Physiol 2007; 293: 119–132. 27 Magalhaes MA, Larson DR, Mader CC, Bravo-Cordero JJ, Gil-Henn H, Oser M et al. 51 Zhang J, Park SI, Artime MC, Summy JM, Shah AN, Bomser JA et al. AFAP-110 is Cortactin phosphorylation regulates cell invasion through a pH-dependent overexpressed in prostate cancer and contributes to tumorigenic growth by pathway. J Cell Biol 2011; 195: 903–920. regulating focal contacts. J Clin Invest 2007; 117: 2962–2973.

Oncogene (2014) 4537 – 4547 & 2014 Macmillan Publishers Limited Substrates of Src AB Reynolds et al 4545 52 Berwanger B, Hartmann O, Bergmann E, Bernard S, Nielsen D, Krause M et al. 77 Kim W, Kook S, Kim DJ, Teodorof C, Song WK. The 31-kDa -generated Loss of a FYN-regulated differentiation and growth arrest pathway in advanced cleavage product of p130cas functions as a transcriptional repressor of E2A in stage neuroblastoma. Cancer Cell 2002; 2: 377–386. apoptotic cells. J Biol Chem 2004; 279: 8333–8342. 53 Reynolds AB, Roesel DJ, Kanner SB, Parsons JT. Transformation-specific tyrosine 78 Wei L, Yang Y, Zhang X, Yu Q. Cleavage of p130Cas in anoikis. J Cell Biochem phosphorylation of a novel cellular protein in chicken cells expressing oncogenic 2004; 91: 325–335. variants of the avian cellular src gene. Mol Cell Biol 1989; 9: 629–638. 79 Casamassima A, Rozengurt E. Tyrosine phosphorylation of p130(cas) by bom- 54 Sakai R, Iwamatsu A, Hirano N, Ogawa S, Tanaka T, Mano H et al. A novel besin, lysophosphatidic acid, phorbol esters, and platelet-derived growth factor. signaling molecule, p130, forms stable complexes in vivo with v-Crk and Signaling pathways and formation of a p130(cas)-Crk complex. J Biol Chem 1997; v-Src in a tyrosine phosphorylation-dependent manner. EMBO J. 1994; 13: 272: 9363–9370. 3748–3756. 80 Casamassima A, Rozengurt E. Insulin-like growth factor I stimulates tyrosine 55 Nakamoto T, Sakai R, Ozawa K, Yazaki Y, Hirai H. Direct binding of C-terminal phosphorylation of p130(Cas), focal adhesion kinase, and paxillin. Role of region of p130Cas to SH2 and SH3 domains of Src kinase. J Biol Chem 1996; 271: phosphatidylinositol 3’-kinase and formation of a p130(Cas).Crk complex. J Biol 8959–8965. Chem 1998; 273: 26149–26156. 56 Pellicena P, Stowell KR, Miller WT. Enhanced phosphorylation of 81 Yamakita Y, Totsukawa G, Yamashiro S, Fry D, Zhang X, Hanks SK et al. Dis- substrates containing SH2 domain binding sites. J Biol Chem 1998; 273: sociation of FAK/p130(CAS)/c-Src complex during mitosis: role of mitosis-specific 15325–15328. serine phosphorylation of FAK. J Cell Biol 1999; 144: 315–324. 57 Pellicena P, Miller WT. Processive phosphorylation of p130Cas by Src depends on 82 Cabodi S, Tinnirello A, Di Stefano P, Bisaro B, Ambrosino E, Castellano I et al. SH3-polyproline interactions. J Biol Chem 2001; 276: 28190–28196. p130Cas as a new regulator of mammary epithelial cell proliferation, survival, 58 Guappone AC, Flynn DC. The integrity of the SH3 binding motif of AFAP-110 is and HER2-neu oncogene-dependent breast tumorigenesis. Cancer Res 2006; 66: required to facilitate tyrosine phosphorylation by, and stable complex formation 4672–4680. with, Src. Mol Cell Biochem 1997; 175: 243–252. 83 van der Flier S, Brinkman A, Look MP, Kok EM, Meijer-van Gelder ME, Klijn JG 59 Guappone AC, Weimer T, Flynn DC. Formation of a stable src-AFAP-110 complex et al. Bcar1/p130Cas protein and primary breast cancer: prognosis and response through either an amino-terminal or a carboxy-terminal SH2-binding motif. Mol to tamoxifen treatment. J Natl Cancer Inst 2000; 92: 120–127. Carcinogen 1998; 22: 110–119. 84 Brinkman A, van der Flier S, Kok EM, Dorssers LC. BCAR1, a human homologue of 60 Sicheri F, Moarefi I, Kuriyan J. Crystal structure of the Src family tyrosine kinase the adapter protein p130Cas, and antiestrogen resistance in breast cancer cells. Hck. Nature 1997; 385: 602–609. J Natl Cancer Inst 2000; 92: 112–120. 61 Williams JC, Weijland A, Gonfloni S, Thompson A, Courtneidge SA, Superti-Furga 85 Dorssers LC, Grebenchtchikov N, Brinkman A, Look MP, van Broekhoven SP, de G et al. The 2.35 a crystal structure of the inactivated form of chicken Src: a Jong D et al. The prognostic value of BCAR1 in patients with primary breast dynamic molecule with multiple regulatory interactions. J Mol Biol 1997; 274: cancer. Clin Cancer Res 2004; 10(18 Pt 1): 6194–6202. 757–775. 86 Fromont G, Vallancien G, Validire P, Levillain P, Cussenot O. BCAR1 expression in 62 Burnham MR, Bruce-Staskal PJ, Harte MT, Weidow CL, Ma A, Weed SA et al. prostate cancer: association with 16q23 LOH status, tumor progression and Regulation of c-SRC activity and function by the adapter protein CAS. Mol Cell EGFR/KAI1 staining. Prostate 2007; 67: 268–273. Biol 2000; 20: 5865–5878. 87 Nick AM, Stone RL, Armaiz-Pena G, Ozpolat B, Tekedereli I, Graybill WS et al. 63 Bouton AH, Riggins RB, Bruce-Staskal PJ. Functions of the adapter protein Cas: Silencing of p130cas in ovarian carcinoma: a novel mechanism for tumor cell signal convergence and the determination of cellular responses. Oncogene 2001; death. J Natl Cancer Inst 2011; 103: 1596–1612. 20: 6448–6458. 88 Huang W, Deng B, Wang RW, Tan QY, He Y, Jiang YG et al. BCAR1 protein plays 64 Cabodi S, del Pilar Camacho-Leal M, Di Stefano P, Defilippi P. Integrin signalling important roles in and predicts poor prognosis in non-small-cell adaptors: not only figurants in the cancer story. Nat Rev Cancer 2010; 10: lung cancer. PLoS One 2012; 7: e36124. 858–870. 89 Riggins RB, Thomas KS, Ta HQ, Wen J, Davis RJ, Schuh NR et al. Physical and 65 Tikhmyanova N, Little JL, Golemis EA. CAS proteins in normal and pathological functional interactions between Cas and c-Src induce tamoxifen resistance of cell growth control. Cell Mol Life Sci 2010; 67: 1025–1048. breast cancer cells through pathways involving 66 Guerrero MS, Parsons JT, Bouton AH. Cas and NEDD9 contribute to tumor pro- receptor and signal transducer and activator of transcription 5b. Cancer Res gression through dynamic regulation of the cytoskeleton. Genes Cancer 2012; 3: 2006; 66: 7007–7015. 371–381. 90 Ta HQ, Thomas KS, Schrecengost RS, Bouton AH. A novel association between 67 Nojima Y, Morino N, Mimura T, Hamasaki K, Furuya H, Sakai R et al. Integrin- p130Cas and resistance to the chemotherapeutic drug adriamycin in human mediated cell adhesion promotes tyrosine phosphorylation of p130Cas, a Src breast cancer cells. Cancer Res 2008; 68: 8796–8804. homology 3-containing molecule having multiple Src homology 2-binding 91 Riggins RB, Quilliam LA, Bouton AH. Synergistic promotion of c-Src activation motifs. J Biol Chem 1995; 270: 15398–15402. and cell migration by Cas and AND-34/BCAR3. J Biol Chem 2003; 278: 68 Harte MT, Hildebrand JD, Burnham MR, Bouton AH, Parsons JT. p130Cas, a 28264–28273. substrate associated with v-Src and v-Crk, localizes to focal adhesions and binds 92 Schuh NR, Guerrero MS, Schrecengost RS, Bouton AH. BCAR3 regulates Src/p130 to focal adhesion kinase. J Biol Chem 1996; 271: 13649–13655. Cas association, Src kinase activity, and breast cancer adhesion signaling. J Biol 69 Honda H, Nakamoto T, Sakai R, Hirai H. p130(Cas), an assembling molecule of Chem 2010; 285: 2309–2317. actin filaments, promotes cell movement, cell migration, and cell spreading in 93 Reynolds AB, Vila J, Lansing TJ, Potts WM, Weber MJ, Parsons JT. Activation of the fibroblasts. Biochem Biophys Res Commun 1999; 262: 25–30. oncogenic potential of the avian cellular src protein by specific structural 70 Harte MT, Macklem M, Weidow CL, Parsons JT, Bouton AH. Identification of two alteration of the carboxy terminus. EMBO J 1987; 6: 2359–2364. focal adhesion targeting sequences in the adapter molecule p130(Cas). Biochim 94 Guan JL, Trevithick JE, Hynes RO. /integrin interaction induces Biophys Acta 2000; 1499: 34–48. tyrosine phosphorylation of a 120-kDa protein. Cell Reg 1991; 2: 71 Meenderink LM, Ryzhova LM, Donato DM, Gochberg DF, Kaverina I, Hanks SK. 951–964. P130Cas Src-binding and substrate domains have distinct roles in sustaining 95 Kornberg LJ, Earp HS, Turner CE, Prockop C, Juliano RL. Signal transduction by focal adhesion disassembly and promoting cell migration. PLoS One 2010; 5: : increased protein tyrosine phosphorylation caused by clustering of e13412. beta 1 integrins. Proc Natl Acad Sci USA 1991; 88: 8392–8396. 72 Nakamoto T, Sakai R, Honda H, Ogawa S, Ueno H, Suzuki T et al. Requirements 96 Golden A, Brugge JS, Shattil SJ. Role of platelet membrane IIb-IIIa in for localization of p130cas to focal adhesions. Mol Cell Biol 1997; 17: 3884–3897. agonist-induced tyrosine phosphorylation of platelet proteins. J Cell Biol 1990; 73 Klemke RL, Leng J, Molander R, Brooks PC, Vuori K, Cheresh DA. CAS/Crk cou- 111(6 Pt 2): 3117–3127. pling serves as a ‘molecular switch’ for induction of cell migration. J Cell Biol 97 Schaller MD, Borgman CA, Cobb BS, Vines RR, Reynolds AB, Parsons JT. 1998; 140: 961–972. pp125FAK a structurally distinctive protein-tyrosine kinase associated with focal 74 Cho SY, Klemke RL. Extracellular-regulated kinase activation and CAS/Crk cou- adhesions. Proc Natl Acad Sci USA 1992; 89: 5192–5196. pling regulate cell migration and suppress apoptosis during invasion of the 98 Parsons JT. Focal adhesion kinase: the first ten years. J Cell Sci 2003; 116(Pt 8): extracellular matrix. J Cell Biol 2000; 149: 223–236. 1409–1416. 75 Cho SY, Klemke RL. Purification of pseudopodia from polarized cells reveals 99 Schaller MD, Hildebrand JD, Shannon JD, Fox JW, Vines RR, Parsons JT. Autop- redistribution and activation of Rac through assembly of a CAS/Crk scaffold. J hosphorylation of the focal adhesion kinase, pp125FAK, directs SH2-dependent Cell Biol 2002; 156: 725–736. binding of pp60src. Mol Cell Biol 1994; 14: 1680–1688. 76 Sawada Y, Tamada M, Dubin-Thaler BJ, Cherniavskaya O, Sakai R, Tanaka S et al. 100 Ren XD, Kiosses WB, Sieg DJ, Otey CA, Schlaepfer DD, Schwartz MA. Focal Force sensing by mechanical extension of the Src family kinase substrate adhesion kinase suppresses Rho activity to promote focal adhesion turnover. p130Cas. Cell 2006; 127: 1015–1026. J Cell Sci 2000; 113(Pt 20): 3673–3678.

& 2014 Macmillan Publishers Limited Oncogene (2014) 4537 – 4547 Substrates of Src AB Reynolds et al 4546 101 Owen KA, Pixley FJ, Thomas KS, Vicente-Manzanares M, Ray BJ, Horwitz AF et al. 126 Provenzano PP, Inman DR, Eliceiri KW, Keely PJ. Matrix density-induced Regulation of lamellipodial persistence, adhesion turnover, and motility in mechanoregulation of breast cell phenotype, signaling and macrophages by focal adhesion kinase. J Cell Biol 2007; 179: 1275–1287. through a FAK-ERK linkage. Oncogene 2009; 28: 4326–4343. 102 Webb DJ, Donais K, Whitmore LA, Thomas SM, Turner CE, Parsons JT et al. FAK- 127 Playford MP, Vadali K, Cai X, Burridge K, Schaller MD. Focal adhesion kinase Src signalling through paxillin, ERK and MLCK regulates adhesion disassembly. regulates cell-cell contact formation in epithelial cells via modulation of Rho. Exp Nat Cell Biol 2004; 6: 154–161. Cell Res 2008; 314: 3187–3197. 103 Ling K, Doughman RL, Firestone AJ, Bunce MW, Anderson RA. Type I gamma 128 Yano H, Mazaki Y, Kurokawa K, Hanks SK, Matsuda M, Sabe H. Roles played by a phosphatidylinositol phosphate kinase targets and regulates focal adhesions. subset of integrin signaling molecules in cadherin-based cell-cell adhesion. JCell Nature 2002; 420: 89–93. Biol 2004; 166: 283–295. 104 Hildebrand JD, Taylor JM, Parsons JT. An SH3 domain-containing GTPase-acti- 129 Avizienyte E, Wyke AW, Jones RJ, McLean GW, Westhoff MA, Brunton VG et al. vating protein for Rho and Cdc42 associates with focal adhesion kinase. Mol Cell Src-induced de-regulation of E-cadherin in colon cancer cells requires integrin Biol 1996; 16: 3169–3178. signalling. Nat Cell Biol 2002; 4: 632–638. 105 Iwanicki MP, Vomastek T, Tilghman RW, Martin KH, Banerjee J, Wedegaertner PB 130 Ilic D, Furuta Y, Kanazawa S, Takeda N, Sobue K, Nakatsuji N et al. Reduced cell et al. FAK, PDZ-RhoGEF and ROCKII cooperate to regulate adhesion movement motility and enhanced focal adhesion contact formation in cells from FAK- and trailing-edge retraction in fibroblasts. J Cell Sci 2008; 121(Pt 6): 895–905. deficient mice. Nature 1995; 377: 539–544. 106 Lim Y, Lim S-T, Tomar A, Gardel M, Bernard-Trifilo JA, Chen XL et al. PyK2 and FAK 131 Furuta Y, Ilic D, Kanazawa S, Takeda N, Yamamoto T, Aizawa S. Mesodermal connections to p190Rho guanine nucleotide exchange factor regulate RhoA defect in late phase of gastrulation by a targeted mutation of focal adhesion activity, focal adhesion formation, and cell motility. J Cell Biol 2008; 180: kinase, FAK. Oncogene 1995; 11: 1989–1995. 187–203. 132 Braren R, Hu H, Kim YH, Beggs HE, Reichardt LF, Wang R. Endothelial FAK is 107 Ren XR, Du QS, Huang YZ, Ao SZ, Mei L, Xiong WC. Regulation of CDC42 GTPase essential for vascular network stability, cell survival, and lamellipodial formation. by proline-rich interacting with PSGAP, a novel pleckstrin J Cell Biol 2006; 172: 151–162. homology and Src homology 3 domain containing rhoGAP protein. J Cell Biol 133 Shen TL, Park AY, Alcaraz A, Peng X, Jang I, Koni P et al. Conditional knockout 2001; 152: 971–984. of focal adhesion kinase in endothelial cells reveals its role in angiogenesis 108 Tomar A, Lim S-T, Lim Y, Schlaepfer DD. A FAK-p120RasGAP-p190RhoGAP and vascular development in late embryogenesis. J Cell Biol 2005; 169: complex regulates polarity in migrating cells. J Cell Sci 2009; 122(Pt 11): 941–952. 1852–1862. 134 Hakim ZS, DiMichele LA, Doherty JT, Homeister JW, Beggs HE, Reichardt LF et al. 109 Chen HC, Appeddu PA, Parsons JT, Hildebrand JD, Schaller MD, Guan JL. Inter- Conditional deletion of focal adhesion kinase leads to defects in ventricular action of focal adhesion kinase with cytoskeletal protein talin. J Biol Chem 1995; septation and outflow tract alignment. Mol Cell Biol 2007; 27: 5352–5364. 270: 16995–16999. 135 Vallejo-Illarramendi A, Zang K, Reichardt LF. Focal adhesion kinase is required for 110 Hildebrand JD, Schaller MD, Parsons JT. Paxillin, a tyrosine phosphorylated focal neural crest cell morphogenesis during mouse cardiovascular development. adhesion-associated protein binds to the carboxyl terminal domain of focal J Clin Invest 2009; 119: 2218–2230. adhesion kinase. Mol Biol Cell 1995; 6: 637–647. 136 Rico B, Beggs HE, Schahin-Reed D, Kimes N, Schmidt A, Reichardt LF. Control of 111 Glenney Jr JR, Zokas L. Novel tyrosine kinase substrates from Rous sarcoma axonal branching and synapse formation by focal adhesion kinase. Nat Neurosci virus-transformed cells are present in the membrane skeleton. J Cell Biol 1989; 2004; 7: 1059–1069. 108: 2401–2408. 137 Forrest AD, Beggs HE, Reichardt LF, Dupree JL, Colello RJ, Fuss B. Focal 112 Lawson C, Lim S-T, Uryu S, Chen XL, Calderwood DA, Schlaepfer DD. FAK pro- adhesion kinase (FAK): a regulator of CNS myelination. J Neurosci Res 2009; 87: motes recruitment of talin to nascent adhesions to control cell motility. J Cell Biol 3456–3464. 2012; 196: 223–232. 138 Nagy T, Wei H, Shen TL, Peng X, Liang CC, Gan B et al. Mammary epithelial- 113 Nayal A, Webb DJ, Horwitz AF. Talin: an emerging focal point of adhesion specific deletion of the focal adhesion kinase gene leads to severe lobulo- dynamics. Curr Opin Cell Biol 2004; 16: 94–98. alveolar hypoplasia and secretory immaturity of the murine mammary gland. 114 Franco SJ, Rodgers MA, Perrin BJ, Han J, Bennin DA, Critchley DR et al. Calpain- J Biol Chem 2007; 282: 31766–31776. mediated proteolysis of talin regulates adhesion dynamics. Nat Cell Biol 2004; 6: 139 Peifer M, Berg S, Reynolds AB. A repeating motif shared by proteins 977–983. with diverse cellular roles. Cell 1994; 76: 789–791. 115 Tomar A, Lawson C, Ghassemian M, Schlaepfer DD. Cortactin as a target for FAK 140 Reynolds AB, Herbert L, Cleveland JL, Berg ST, Gaut JR. p120, a novel substrate of in the regulation of focal adhesion dynamics. PLoS One 2012; 7: 8 e44041. 116 Ezratty EJ, Partridge MA, Gundersen GG. Microtubule-induced focal adhesion protein tyrosine kinase receptors and of p60v-src, is related to cadherin-binding disassembly is mediated by dynamin and focal adhesion kinase. Nat Cell Biol factors beta-catenin, plakoglobin and armadillo. Oncogene 1992; 7: 2439–2445. 2005; 7: 581–590. 141 Thoreson MA, Anastasiadis PZ, Daniel JM, Ireton RC, Wheelock MJ, Johnson KR 117 Wang Y, Cao H, Chen J, McNiven MA. A direct interaction between the large et al. Selective uncoupling of p120(ctn) from E-cadherin disrupts strong adhe- GTPase dynamin-2 and FAK regulates focal adhesion dynamics in response to sion. J Cell Biol 2000; 148: 189–202. active Src. Mol Biol Cell 2011; 22: 1529–1538. 142 Reynolds AB, Daniel J, McCrea PD, Wheelock MJ, Wu J, Zhang Z. Identification of 118 Beggs HE, Schahin-Reed D, Zang K, Goebbels S, Nave KA, Gorski J et al. a new catenin: the tyrosine kinase substrate p120cas associates with E-cadherin FAK deficiency in cells contributing to the basal lamina results in cortical complexes. Mol Cell Biol 1994; 14: 8333–8342. abnormalities resembling congenital muscular dystrophies. Neuron 2003; 40: 143 Dillon DA, D’Aquila T, Reynolds AB, Fearon ER, Rimm DL. The expression of 501–514. p120ctn protein in breast cancer is independent of alpha- and beta-catenin and 119 Midwood KS, Schwarzbauer JE. -C modulates matrix contraction via E-cadherin. Am J Pathol 1998; 152: 75–82. focal adhesion kinase- and Rho-mediated signaling pathways. Mol Biol Cell 2002; 144 Anastasiadis PZ, Moon SY, Thoreson MA, Mariner DJ, Crawford HC, Zheng Y et al. 13: 3601–3613. Inhibition of RhoA by p120 catenin. Nat Cell Biol 2000; 2: 637–644. 120 Hauck CR, Sieg DJ, Hsia DA, Loftus JC, Gaarde WA, Monia BP et al. Inhibition of 145 Anastasiadis PZ, Reynolds AB. Regulation of Rho GTPases by p120-catenin. focal adhesion kinase expression or activity disrupts epidermal growth factor- Curr Opin Cell Biol 2001; 13: 604–610. stimulated signaling promoting the migration of invasive human carcinoma 146 Birukova AA, Zebda N, Cokic I, Fu P, Wu T, Dubrovskyi O et al. p190RhoGAP cells. Cancer Res 2001; 61: 7079–7090. mediates protective effects of oxidized phospholipids in the models of venti- 121 Wu X, Gan B, Yoo Y, Guan J-L. FAK-mediated src phosphorylation of endophilin lator-induced lung injury. Exp Cell Res 2011; 317: 859–872. A2 inhibits endocytosis of MT1-MMP and promotes ECM degradation. Dev Cell 147 Ponik SM, Trier SM, Wozniak MA, Eliceiri KW, Keely PJ. RhoA is down-regulated at 2005; 9: 185–196. cell-cell contacts via p190RhoGAP-B in response to tensional homeostasis. Mol 122 Pan Y-R, Chen C-L, Chen H-C. FAK is required for the assembly of podosome Biol Cell 2013; 24: 1688–1699. rosettes. J Cell Biol 2011; 195: 113–129. 148 Wildenberg GA, Dohn MR, Carnahan RH, Davis MA, Lobdell NA, Settleman J et al. 123 Wang Y, McNiven MA. Invasive matrix degradation at focal adhesions occurs p120-catenin and p190RhoGAP regulate cell-cell adhesion by coordinating via protease recruitment by a FAK-p130Cas complex. J Cell Biol 2012; 196: antagonism between Rac and Rho. Cell 2006; 127: 1027–1039. 375–385. 149 Smith AL, Dohn MR, Brown MV, Reynolds AB. Association of Rho-associated 124 Klein EA, Yin L, Kothapalli D, Castagnino P, Byfield FJ, Xu T et al. Cell-cycle control protein kinase 1 with E-cadherin complexes is mediated by p120-catenin. by physiological matrix elasticity and in vivo tissue stiffening. Curr Biol 2009; 19: Mol Biol Cell 2012; 23: 99–9110. 1511–1518. 150 Miyashita Y, Ozawa M. Increased internalization of p120-uncoupled E-cadherin 125 Parekh A, Weaver AM. Regulation of cancer invasiveness by the physical extra- and a requirement for a dileucine motif in the cytoplasmic domain for endo- cellular matrix environment. Cell Adh Migr 2009; 3: 288–292. cytosis of the protein. J Biol Chem 2007; 282: 11540–11548.

Oncogene (2014) 4537 – 4547 & 2014 Macmillan Publishers Limited Substrates of Src AB Reynolds et al 4547 151 Sato K, Watanabe T, Wang S, Kakeno M, Matsuzawa K, Matsui T et al. Numb 170 Manley PW, Drueckes P, Fendrich G, Furet P, Liebetanz J, Martiny-Baron G et al. controls E-cadherin endocytosis through p120 catenin with aPKC. Mol Biol Cell Extended kinase profile and properties of the protein kinase inhibitor nilotinib. 2011; 22: 3103–3119. Biochim Biophys Acta 2010; 1804: 445–453. 152 Kanner SB, Reynolds AB, Parsons JT. Tyrosine phosphorylation of a 120-kilo- 171 Parsons SJ, Parsons JT. Src family kinases, key regulators of signal transduction. dalton pp60src substrate upon epidermal growth factor and platelet-derived Oncogene 2004; 23: 7906–7909. growth factor receptor stimulation and in polyomavirus middle-T-antigen- 172 Frame MC. Newest findings on the oldest oncogene; how activated src does it. J transformed cells. Mol Cell Biol 1991; 11: 713–720. Cell Sci 2004; 117(Pt 7): 989–998. 153 Downing JR, Reynolds AB. PDGF, CSF-1, and EGF induce tyrosine phosphoryla- 173 Gallick GE, Corn PG, Zurita AJ, Lin SH. Small-molecule protein tyrosine kinase tion of p120, a pp60src transformation-associated substrate. Oncogene 1991; 6: inhibitors for the treatment of metastatic prostate cancer. Future Med Chem 607–613. 2012; 4: 107–119. 154 Rodriguez FJ, Lewis-Tuffin LJ, Anastasiadis PZ. E-cadherin’s dark side: possible 174 Kopetz S, Shah AN, Gallick GE. Src continues aging: current and future clinical role in tumor progression. Biochim Biophys Acta 2012; 1826: 23–31. directions. Clin Cancer Res 2007; 13: 7232–7236. 155 Smalley-Freed WG, Efimov A, Burnett PE, Short SP, Davis MA, Gumucio DL et al. 175 Tilghman RW, Parsons JT. Focal adhesion kinase as a regulator of cell tension in p120-catenin is essential for maintenance of barrier function and intestinal the progression of cancer. Semin Cancer Biol 2008; 18: 45–52. homeostasis in mice. J Clin Invest 2010; 120: 1824–1835. 176 Tomar A, Schlaepfer DD. Focal adhesion kinase: switching between GAPs and 156 Smalley-Freed WG, Efimov A, Short SP, Jia P, Zhao Z, Washington MK et al. GEFs in the regulation of cell motility. Curr Opin Cell Biol 2009; 21: 676–683. Adenoma formation following limited ablation of p120-catenin in the mouse 177 Schaller MD. Cellular functions of FAK kinases: insight into molecular mechan- intestine. PLoS One 2011; 6: e19880. isms and novel functions. J Cell Sci 2010; 123(Pt 7): 1007–1013. 157 Perez-Moreno M, Davis MA, Wong E, Pasolli HA, Reynolds AB, Fuchs E. p120- 178 Zhao X, Guan J-L. Focal adhesion kinase and its signaling pathways in cell catenin mediates inflammatory responses in the skin. Cell 2006; 124: 631–644. migration and angiogenesis. Adv Drug Deliv Rev 2011; 63: 610–615. 158 Stairs DB, Bayne LJ, Rhoades B, Vega ME, Waldron TJ, Kalabis J et al. Deletion of 179 Han EK-H, McGonigal T. Role of focal adhesion kinase in human cancer: a p120-catenin results in a tumor microenvironment with inflammation and potential target for drug discovery. Anticancer Agents Med Chem 2007; 7: cancer that establishes it as a tumor suppressor gene. Cancer Cell 2011; 19: 681–684. 470–483. 180 Schwock J, Dhani N, Hedley DW. Targeting focal adhesion kinase signaling in 159 Kurley SJ, Bierie B, Carnahan RH, Lobdell NA, Davis MA, Hofmann I et al. p120- tumor growth and metastasis. Expert Opin Ther Targets 2010; 14: 77–94. catenin is essential for terminal end bud function and mammary morphogen- 181 Ma WW. Development of focal adhesion kinase inhibitors in cancer therapy. esis. Development 2012; 139: 1754–1764. Anticancer Agents Med Chem 2011; 11: 638–642. 160 Davis MA, Reynolds AB. Blocked acinar development, E-cadherin reduction, and 182 Schultze A, Fiedler W. Clinical importance and potential use of small molecule intraepithelial neoplasia upon ablation of p120-catenin in the mouse salivary inhibitors of focal adhesion kinase. Anticancer Agents Med Chem 2011; 11: gland. Dev Cell 2006; 10: 21–31. 593–599. 161 Thoreson MA, Reynolds AB. Altered expression of the catenin p120 in human 183 Dunn KB, Heffler M, Golubovskaya VM. Evolving therapies and FAK inhibitors for cancer: implications for tumor progression. Differentiation 2002; 70: 583–589. the treatment of cancer. Anticancer Agents Med Chem 2010; 10: 722–734. 162 David E, Tramontin T, Zemmel R. Pharmaceutical R&D: the road to positive 184 Reynolds AB, Roczniak-Ferguson A. Emerging roles for p120-catenin in cell returns. Nat Rev Drug Discov 2009; 8: 609–610. adhesion and cancer. Oncogene 2004; 23: 7947–7956. 163 Paul SM, Mytelka DS, Dunwiddie CT, Persinger CC, Munos BH, Lindborg SR et al. 185 Reynolds AB. p120-catenin: past and present. Biochim Biophys Acta. 2007; 1773: How to improve R&D productivity: the pharmaceutical industry’s grand chal- 2–7. lenge. Nat Rev Drug Discov 2010; 9: 203–214. 186 Ammer AG, Weed SA. Cortactin branches out: roles in regulating protrusive actin 164 Berggren R, Moller M, Moss R, Poda P, Smietana K. Outlook for the next 5 years in dynamics. Cell Motil Cytoskeleton 2008; 65: 687–707. drug innovation. Nat Rev Drug Discov 2012; 11: 435–436. 187 Kirkbride KC, Sung BH, Sinha S, Weaver AM. Cortactin: a multifunctional regulator 165 Brunton VG, Frame MC. Src and focal adhesion kinase as therapeutic targets in of cellular invasiveness. Cell Adh Migr 2011; 5: 187–198. cancer. Curr Opin Pharmacol 2008; 8: 427–432. 188 Wells JA, McClendon CL. Reaching for high-hanging fruit in drug discovery at 166 Creedon H, Brunton VG. Src kinase inhibitors: promising cancer therapeutics? Crit protein-protein interfaces. Nature 2007; 450: 1001–1009. Rev Oncog 2012; 17: 145–159. 189 Khan SH, Ahmad F, Ahmad N, Flynn DC, Kumar R. Protein-protein interactions: 167 Abbott BL. Dasatinib: from treatment of -resistant or -intolerant patients principles, techniques, and their potential role in new drug development. with chronic myeloid leukemia to treatment of patients with newly diagnosed J Biomol Struct Dyn 2011; 28: 929–938. chronic phase chronic myeloid leukemia. Clin Ther 2012; 34: 272–281. 190 Radi M, Schenone S, Botta M. Allosteric inhibitors of Bcr-Abl: towards novel 168 Quintas-Cardama A, Kantarjian H, Cortes J. Bosutinib for the treatment of chronic myristate-pocket binders. Curr Pharm Biotechnol. (e-pub ahead of print 20 March myeloid leukemia in chronic phase. Drugs Today 2012; 48: 177–188. 2012). 169 Hantschel O, Grebien F, Superti-Furga G. The growing arsenal of ATP-competi- 191 Hopkins AL. Network pharmacology: the next paradigm in drug discovery. tive and allosteric inhibitors of BCR-ABL. Cancer Res 2012; 72: 4890–4895. Nat Chem Biol 2008; 4: 682–690.

& 2014 Macmillan Publishers Limited Oncogene (2014) 4537 – 4547