11
Commentary 1183 Introduction Knowledge of cell adhesion proteins and the molecules that associate with them has grown rapidly in recent years. Cadherins and integrins are involved in both bidirectional cell signaling events, and the physical linkages of cells to each other and to the extracellular matrix (ECM). Physical adhesive linkages are crucial for the maintenance of tissue architecture and can also serve instructive roles by enabling cells to sense and respond to changes in their environments. In some cases, this occurs through translation of mechanical inputs into intracellular signals, a process known as mechanotransduction. A simple survey of adhesion-dependent cell signaling pathways reveals that many of the molecular components and functional outputs are common to several different types of adhesion. This leads to questions of how and where these signaling pathways intersect, and what functional consequences result from these interactions? Although other adhesion molecules are likely to be involved and can be viewed as additional nodes in an overall cellular adhesive network, this review focuses on integrin-based cell–ECM interactions and cadherin-dependent cell–cell contacts because a more coherent picture of interactions between these functionally important adhesions is now emerging. Adhesive crosstalk – re-evaluation of concepts and the case for adhesive networks The term crosstalk is typically used to represent an interaction(s) between two or more independently initiated signaling pathways, the outcomes of which include the amplification or attenuation of individual pathways, or the initiation of new signals. In the context of signals transduced through integrins and cadherins these pathways intersect in ways that resemble more closely an integrated network rather than distinct cascades (see Box 1). Integrins and cadherins are both transmembrane adhesion receptors, have many signaling effector molecules in common, link to common scaffolding and cytoskeletal elements, and share the ability to influence crucial downstream functions, such as cell growth, survival and transcriptional activity. Owing to these common features and molecular associations, cell signaling pathways that depend on cadherins and integrins are likely to interact on multiple levels, and these interactions occur over varying time and length scales. We can distinguish such networks on the basis of both short- and long-range physical associations and cell signaling events. By these criteria we define three general modes of adhesive interactions (Fig. 1). In the most-indirect or -remote mode of adhesive interactions, signals that originate from one type of adhesion lead to a change in the functional activity of other adhesive contacts elsewhere in the cell (Fig. 1A). We term this ‘long-range input–output’ signaling. For example, cell adhesion to specific ECM proteins or to neighboring cells might lead to changes in gene expression, which could include alterations in levels of adhesion molecules or other proteins involved in regulating adhesion (Onodera et al., 2010). Alternatively, engagement or disengagement of one type of adhesion might modify the functional activities of another by effecting changes in membrane trafficking, cytoskeletal association and/or avidity or binding affinity (Avizienyte et al., 2002). Another type of interaction between adhesions involves the convergence of independently initiated cell signaling events, often involving downstream effectors that are common to both integrin and cadherin adhesions (Fig. 1B). These shared effectors include non-receptor tyrosine kinases, adaptor and scaffolding proteins, and small GTPases. In addition, cell–cell and cell–matrix adhesions are also linked to the common structural elements that comprise the cytoskeleton. Actin, microtubules and intermediate filaments form distinct, but often spatially overlapping, macromolecular Summary Cell–cell and cell–extracellular-matrix (cell–ECM) adhesions have much in common, including shared cytoskeletal linkages, signaling molecules and adaptor proteins that serve to regulate multiple cellular functions. The term ‘adhesive crosstalk’ is widely used to indicate the presumed functional communication between distinct adhesive specializations in the cell. However, this distinction is largely a simplification on the basis of the non-overlapping subcellular distribution of molecules that are involved in adhesion and adhesion-dependent signaling at points of cell–cell and cell–substrate contact. The purpose of this Commentary is to highlight data that demonstrate the coordination and interdependence of cadherin and integrin adhesions. We describe the convergence of adhesive inputs on cell signaling pathways and cytoskeletal assemblies involved in regulating cell polarity, migration, proliferation and survival, differentiation and morphogenesis. Cell–cell and cell–ECM adhesions represent highly integrated networks of protein interactions that are crucial for tissue homeostasis and the responses of individual cells to their adhesive environments. We argue that the machinery of adhesion in multicellular tissues comprises an interdependent network of cell–cell and cell–ECM interactions and signaling responses, and not merely crosstalk between spatially and functionally distinct adhesive specializations within cells. Key words: Adhesion, Cadherin, Crosstalk, Integrin, Mechanotransduction Journal of Cell Science 124, 1183-1193 © 2011. Published by The Company of Biologists Ltd doi:10.1242/jcs.064618 Integrins and cadherins join forces to form adhesive networks Gregory F. Weber, Maureen A. Bjerke and Douglas W. DeSimone* Department of Cell Biology, School of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA *Author for correspondence ([email protected]) Journal of Cell Science

Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

Commentary 1183

IntroductionKnowledge of cell adhesion proteins and the molecules thatassociate with them has grown rapidly in recent years. Cadherinsand integrins are involved in both bidirectional cell signalingevents, and the physical linkages of cells to each other and to theextracellular matrix (ECM). Physical adhesive linkages are crucialfor the maintenance of tissue architecture and can also serveinstructive roles by enabling cells to sense and respond to changesin their environments. In some cases, this occurs through translationof mechanical inputs into intracellular signals, a process known asmechanotransduction. A simple survey of adhesion-dependent cellsignaling pathways reveals that many of the molecular componentsand functional outputs are common to several different types ofadhesion. This leads to questions of how and where these signalingpathways intersect, and what functional consequences result fromthese interactions? Although other adhesion molecules are likely tobe involved and can be viewed as additional nodes in an overallcellular adhesive network, this review focuses on integrin-basedcell–ECM interactions and cadherin-dependent cell–cell contactsbecause a more coherent picture of interactions between thesefunctionally important adhesions is now emerging.

Adhesive crosstalk – re-evaluation of conceptsand the case for adhesive networksThe term crosstalk is typically used to represent an interaction(s)between two or more independently initiated signaling pathways,the outcomes of which include the amplification or attenuation ofindividual pathways, or the initiation of new signals. In the contextof signals transduced through integrins and cadherins thesepathways intersect in ways that resemble more closely an integratednetwork rather than distinct cascades (see Box 1). Integrins andcadherins are both transmembrane adhesion receptors, have many

signaling effector molecules in common, link to commonscaffolding and cytoskeletal elements, and share the ability toinfluence crucial downstream functions, such as cell growth,survival and transcriptional activity. Owing to these commonfeatures and molecular associations, cell signaling pathways thatdepend on cadherins and integrins are likely to interact on multiplelevels, and these interactions occur over varying time and lengthscales. We can distinguish such networks on the basis of bothshort- and long-range physical associations and cell signalingevents. By these criteria we define three general modes of adhesiveinteractions (Fig. 1).

In the most-indirect or -remote mode of adhesive interactions,signals that originate from one type of adhesion lead to a changein the functional activity of other adhesive contacts elsewhere inthe cell (Fig. 1A). We term this ‘long-range input–output’ signaling.For example, cell adhesion to specific ECM proteins or toneighboring cells might lead to changes in gene expression, whichcould include alterations in levels of adhesion molecules or otherproteins involved in regulating adhesion (Onodera et al., 2010).Alternatively, engagement or disengagement of one type ofadhesion might modify the functional activities of another byeffecting changes in membrane trafficking, cytoskeletal associationand/or avidity or binding affinity (Avizienyte et al., 2002).

Another type of interaction between adhesions involves theconvergence of independently initiated cell signaling events, ofteninvolving downstream effectors that are common to both integrinand cadherin adhesions (Fig. 1B). These shared effectors includenon-receptor tyrosine kinases, adaptor and scaffolding proteins,and small GTPases. In addition, cell–cell and cell–matrix adhesionsare also linked to the common structural elements that comprisethe cytoskeleton. Actin, microtubules and intermediate filamentsform distinct, but often spatially overlapping, macromolecular

SummaryCell–cell and cell–extracellular-matrix (cell–ECM) adhesions have much in common, including shared cytoskeletal linkages, signalingmolecules and adaptor proteins that serve to regulate multiple cellular functions. The term ‘adhesive crosstalk’ is widely used toindicate the presumed functional communication between distinct adhesive specializations in the cell. However, this distinction islargely a simplification on the basis of the non-overlapping subcellular distribution of molecules that are involved in adhesion andadhesion-dependent signaling at points of cell–cell and cell–substrate contact. The purpose of this Commentary is to highlight datathat demonstrate the coordination and interdependence of cadherin and integrin adhesions. We describe the convergence of adhesiveinputs on cell signaling pathways and cytoskeletal assemblies involved in regulating cell polarity, migration, proliferation and survival,differentiation and morphogenesis. Cell–cell and cell–ECM adhesions represent highly integrated networks of protein interactions thatare crucial for tissue homeostasis and the responses of individual cells to their adhesive environments. We argue that the machineryof adhesion in multicellular tissues comprises an interdependent network of cell–cell and cell–ECM interactions and signalingresponses, and not merely crosstalk between spatially and functionally distinct adhesive specializations within cells.

Key words: Adhesion, Cadherin, Crosstalk, Integrin, Mechanotransduction

Journal of Cell Science 124, 1183-1193 © 2011. Published by The Company of Biologists Ltddoi:10.1242/jcs.064618

Integrins and cadherins join forces to form adhesivenetworksGregory F. Weber, Maureen A. Bjerke and Douglas W. DeSimone*Department of Cell Biology, School of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA*Author for correspondence ([email protected])

Jour

nal o

f Cel

l Sci

ence

Page 2: Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

assemblies. These cytoskeletal networks provide physical scaffoldsthat connect adhesion complexes not only proximally but also at adistance across cells and cell junctions.

The lateral coupling of adhesion receptors can be viewed as athird form of interaction, which involves more short-rangeassociations within the plane of the membrane (Fig. 1C). In thisinstance, however, the proximal interactions of integrin andcadherin do not necessarily involve shared cytoskeletal linkages,or even cell–cell or cell–ECM engagement. Adaptor proteins, suchas tetraspanins or growth factor receptors – e.g. the insulin-likegrowth factor 1 receptor (IGF1R) – can facilitate lateral associationsof integrins and cadherins (Chattopadhyay et al., 2003; Canoniciet al., 2008). One result of this type of interaction is that integrinspromote stability of cell–cell adhesions (Chattopadhyay et al.,2003). Although lateral integrin–cadherin associations are knownto occur, their physiological significance remains unclear.

Each of these three modes of adhesive interaction can alsoconverge on a common pathway(s), resulting in a complex feedbackloop that, in turn, modulates the functions of one or more of theinitiating signals (Fig. 1D). For example, RhoGTPases act as bothpoints of convergent signaling downstream of adhesions as well asupstream modifiers of functions of individual adhesion molecules.In the following sections we consider examples of adhesivenetworking in the context of the three types of general mechanismthat are illustrated in Fig. 1A-C and Table 1.

Mechanisms for integrating adhesive signals RhoGTPasesRhoGTPases are central to cell signaling pathways both upstreamand downstream of cadherin and integrin adhesions (Huveneersand Danen, 2009; Watanabe et al., 2009), making them primecandidates for mediating integration of adhesion-dependent signals.They are known to modulate a wide range of cellular behaviorsincluding cytoskeletal organization, cell polarity, cell proliferation,and the formation and maturation of adhesive junctions (Etienne-Manneville and Hall, 2002). RhoGTPases have a primary role inregulating the assembly of integrin-based focal adhesions (Hall,1998). Similarly, the assembly of cadherin-based adherens junctions

requires the activity of Rho, Rac and Cdc42 (Van Aelst and Symons,2002). Rho GTPases must be tightly regulated in the cell given thata certain degree of Rho activity is required for cell–cell adhesionbut increased levels of Rho activity disrupt cadherin adhesions(Zhong et al., 1997).

Adhesion through classical cadherins results in increased Rac1activity upon engagement but inhibits RhoA activity over thecourse of a few hours (Noren et al., 2001). The effects of cadherinadhesion on RhoGTPases are known to require the cytoplasmicdomain of cadherin, but the mechanism of signal transductionremains to be established (Noren et al., 2001; Watanabe et al.,2009). A number of proteins bind the cytoplasmic tails of cadherinsand some of these have been shown to regulate RhoGTPase activity.p120 catenin binds to the juxtamembrane region of the cadherintail and, when overexpressed, can decrease Rho activity whileincreasing Rac and Cdc42 activity (Braga and Yap, 2005). Anotherstudy suggested that p120 catenin interacts with p190 Rho GTPaseactivating protein (RhoGAP), locally inhibiting Rho in response to

1184 Journal of Cell Science 124 (8)

Shared signaling effectorproteins (e.g. Rho GTPases)

Remote regulation of expression or activity(e.g. suppression of cadherin expression)

A Input–output signaling C Lateral coupling

B Convergent signaling

Adaptor orscaffolding

proteins

Shared signalingeffector proteins

D Convergent signaling with long-range feedback

Downstreamsignaling effects

Downstreamsignaling effects

Downstream signalingcascade

Fig. 1. Different modes of adhesive interaction. (A)Input–output signalingis used to describe long-range interactions between cadherins (monomers inblue) and integrins (heterodimers in green). Signaling from one adhesionreceptor modulates the expression or activity of another adhesion type. Theunderlying mechanisms may involve changes in transcriptional activity orchanges in signaling effector activity that regulate cell–cell and cell–matrixadhesions. (B)Convergent signaling occurs when cadherins and integrinssignal to common downstream effector molecules. These shared effectorsinclude kinases, adaptor proteins and cytoskeletal components. Convergentsignaling allows for cadherins and integrins to jointly affect cell physiologythrough redundant, additive or synergistic mechanisms. (C)Lateral coupling ofadhesion receptors allows for signaling in the absence of extracellular matrixor cell–cell adhesion. Macromolecular complexes are formed by association ofcadherins with integrins, often through interaction with other transmembraneproteins, such as tetraspanins (orange) or growth factor receptors (purple).Cytoplasmic adaptor or scaffolding proteins (pink oval), such as -catenintogether with cytoskeletal connections, can further support association of thesemolecular complexes. (D)All three modes shown in A–C may work inconjunction with one another.

Box 1. Adhesive networksThe term ‘adhesive crosstalk’ is used widely to highlightpresumed functional interactions between two distinct types ofadhesion (e.g. integrin and cadherin adhesions; see Figure A,blue and green dots, respectively). Although often spatiallydistinct, integrin and cadherin adhesions activate many of thesame signaling pathways and elicit similar cellular functions,supporting the notion that they should instead be considered asinterdependent functional nodes in a larger adhesive network(see Figure B). In some cellular contexts, cadherins and integrinsare best perceived as functionally equivalent (teal nodes) withrespect to output and network response. Modulation of one nodeinfluences adhesive function and signaling activities of adhesivenodes throughout the network, symbolized by the simple graphicrepresentation.

A Simple crosstalk B Integrated network

Jour

nal o

f Cel

l Sci

ence

Page 3: Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

Rac activation (Wildenberg et al., 2006). p190 RhoGAP is activatedby integrin adhesion and is thought to be a convergence point forsignaling by integrins and syndecans (Bass et al., 2008). In epitheliaand endothelia, in which both cell–cell and cell–matrix adhesionsare required, p190 RhoGAP is likely to be a point of convergentsignaling between integrins and cadherins.

Rho is transiently activated by the formation of new integrinadhesions (Ren et al., 1999; Bhadriraju et al., 2007), and thisactivation can have consequences for the adhesive functions ofcadherins. There are several examples of RhoGTPases that act assignaling intermediaries between integrins and cadherins. Theactivities of Rho and Rac downstream of integrin signaling arethought to regulate the formation of adherens junctions in epithelialcells (Playford et al., 2008). Integrin signaling in colon cancer cellspromotes cell–cell junction formation through activation ofphosphatidylinositol 3-kinase (PI 3-kinase) and Rac1B (Chartier etal., 2006). In addition to these input–output pathways, RhoGTPasesare also implicated in convergent signaling. For example, bothintegrin and cadherin adhesions have been shown to enhance cellproliferation by promoting the expression of cyclin D1 in aredundant manner through Rac (Fig. 2) (Fournier et al., 2008).

Rho activation is a node in the convergent signaling networkinitiated by cadherins and integrins that can exert varied effects onadhesions depending on the downstream effectors involved (Fig. 2).Rho signaling through diaphamous Dia reorganizes the actincytoskeleton to stabilize adherens junctions, whereas Rho-kinase(ROCK) is thought to disrupt cell–cell junctions by activatingactomyosin contractility to excess (Sahai and Marshall, 2002).

RhoA-mediated activation of the effector ROCK is regulated bycell–matrix adhesion, cell shape and cytoskeletal tension. In fact,tension generated by cell spreading is required for activation ofROCK by RhoA (Bhadriraju et al., 2007). This suggests theexistence of a positive feedback loop, in which tension generatedby cell–matrix adhesion stimulates activation of ROCK, which inturn enhances the formation and maturation of integrin-basedadhesions. Regulation of cytoskeletal tension is also crucial for theaccumulation of E-cadherin at cell–cell contacts. ROCK regulatesthe activity of the motor protein nonmuscle myosin II downstreamof initial E-cadherin ligation in order to stabilize newly formedcell–cell junctions (Shewan et al., 2005). Cdc42 limits Rhosignaling to achieve the crucial tension levels that are required tomaintain cell–cell junctions, preventing excess tension that wouldprobably lead to the dissociation of these adhesions (Warner andLongmore, 2009).

Owing to the complex spatiotemporal regulation of RhoGTPases,it is difficult to separate their roles in the initial establishmentof adhesions from those in the signaling events downstream ofcadherin and integrin engagement. Furthermore, because theseproteins are involved in so many pathways, localization and timingbecome extremely important in dictating cellular responses to agiven stimulus. Rac1 is localized to sites of cell–cell contact whereit might have a role in mediating rapid changes in actin organizationconcurrent with the formation of new cell–cell junctions (Ehrlichet al., 2002). More recent work has shown that localization of bothRac and Rho to cell–cell contacts is essential for the formation andexpansion of cadherin adhesions. The spatiotemporal localization

1185Adhesive crosstalk

Table 1. Examples of interactions between cadherins and integrins

Effector Type of signaling Cellular or Cadherin Integrin intermediate interaction Cell or tissue type physiological condition Citation

E-cadherin 1-integrin Rac1, cyclin D1 Convergent MCF10A mammary Proliferation (Fournier et al., 2008)epithelial

N-cadherin 1-integrin PTP1B Convergent PC12, chick neural Neurite outgrowth (Pathre et al., 2001)retina explants

DE-cadherin -integrin Rac, JNK Convergent, Drosophila border cells Collective cell migration (Llense and Martin-input–output Blanco, 2008; Wang

et al., 2010)N-cadherin 1-integrin Fer Convergent, Chick neural retina Neurite outgrowth (Arregui et al., 2000)

input–output explantsE-cadherin 51-integrin Btbd7 Input–output Salivary gland Branching (Sakai et al., 2003;

morphogenesis Onodera et al., 2010)E-cadherin v- or Src, ROCK or Input–output L fibroblasts, S180 Cell–cell adhesion (Martinez-Rico et al.,

51-integrin MLCK mouse sarcoma, SCC13 2010)squamous cell carcinoma

E-cadherin 1-integrin FAK Input–output Human colon carcinoma Epithelial–mesenchymal (Wang et al., 2004)Moser cells transition

E-cadherin Collagen receptor Matrix Input–output Ovarian carcinoma cells Epithelial–mesenchymal (Symowicz et al.,metalloproteinase transition 2007)

E-cadherin Fibronectin receptor Rap1 Input–output Fisher rat thyroid (FRT) Cell–matrix adhesion (Balzac et al., 2005)cells

E-cadherin Laminin receptor Rac1b, PI 3kinase Input–output HT-29 human colon Cell–cell adhesion (Chariter et al., 2006)adenocarcinoma

N-cadherin 1- and 3-integrin Ca2+ Input–output Neural crest cells Cell migration (Monier-Gavelle andDuband, 1997,Theveneau et al.,2010)

N-cadherin Fibronectin receptor Rac1 Input–output Human mesenchymal Myogenesis (Gao et al., 2010)stem cells

VE-cadherin v3-integrin VEGFR2, Shc Input–output Bovine aortic endothelial Inflammation (Tzima et al., 2005;cells Liu et al., 2008)

E-cadherin 31-integrin CD151 Lateral coupling Immortalized mouse Cell–cell adhesion (Chattopadhyay et al.,kidney epithelia 2003)

E-cadherin v-integrin IGF-1R, -catenin Lateral coupling Human colonic Cell migration (Canonici et al., 2008)adenocarcinoma

Jour

nal o

f Cel

l Sci

ence

Page 4: Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

of both the GTPases and their effectors suggests specific roles inadhesion formation, with Rac facilitating actin remodeling andRho stimulating actomyosin contractility (Yamada and Nelson,2007). The downstream effects of RhoGTPase activation can differdramatically depending on the site of activation. Rac is requiredfor epithelial wound healing and promotes cell migration whenactivated at cell–matrix contact sites, but promotes cell–celladhesion and formation of adherens junctions when activated atcell–cell junctions (Van Aelst and Symons, 2002; Liu et al., 2010).Network complexity increases when we consider the functions ofother GTPases, such as the Ras family member Rap1, which hasbeen implicated in regulation of integrin activity downstream ofcadherins (see Box 2). Clearly, signaling through GTPases is amuch-used mechanism for intracellular communication, and isinfluenced by both cell–cell and cell–ECM adhesions. Only recentlywere suitable tools developed that enable GTPase activities to bevisualized in time and space, thus revealing specific activationevents that mediate interactions between adhesions (Yamada andNelson, 2007; Wang et al., 2010).

Tyrosine kinasesA number of tyrosine kinases are localized to cell–cell and cell–matrix adhesions (Giannone and Sheetz, 2006; McLachlan et al.,2007) where they function as prominent nodes in the adhesivenetwork. Activation of Src family kinases frequently accompaniesthe formation of both cell–cell and cell–ECM adhesions. Src isrecruited and activated upon E-cadherin ligation and this provides

a positive feedback loop that signals through PI 3-kinase to promotethe stability of cell–cell contacts (McLachlan et al., 2007). However,Src levels at cell–cell adhesions must be tightly regulated, becauseconstitutively active Src disrupts cell–cell contacts and alters cellmorphology (Behrens et al., 1993). Integrin ligation also leads toSrc activation and this has divergent downstream consequences,often involving RhoGTPases (Huveneers and Danen, 2009). Inepithelial cells, constitutively active Src at sites of integrin–matrixadhesion leads to peripheral accumulation of activated myosin,which is disruptive to cell–cell junctions (Avizienyte et al., 2004).However, moderate Src activation and regulation of actomyosincontractility by ROCK or myosin light-chain kinase (MLCK) arenecessary for the integrin-mediated strengthening of E-cadherinadhesions (Martinez-Rico et al., 2010). Although cytoskeletaltension is important for the formation of both cell–cell and cell–matrix adhesions, excessive tension might serve to rip junctionsapart or induce changes in protein conformation that lead tojunctional instability.

The focal adhesion kinase (Fak, also known as PTK2) is anothernon-receptor tyrosine kinase, which as a primary signaling partnerof Src is implicated in many of the same signaling pathways(Playford and Schaller, 2004). Unlike Src, however, Fak containsa focal adhesion targeting (FAT) sequence, consistent with its roleas a downstream effector of integrin adhesion and signaling. Forexample, transforming growth factor- (TGF-) inhibits epithelial-to-mesenchymal transition (EMT) in at least some colon cancercells, and stimulates increased expression of ECM leading to

1186 Journal of Cell Science 124 (8)

Cell migration

Shear stress

Inflammation

Cell–cell junction dissolution

Cell–cell adhesion

Cell–ECM adhesion

Cell–cell adhesion

Cyclic strain?

Proliferation

ShcP

FerP

CortactinP

NF-κBERK

P

ShcP

RhoGTP

ROCKRho

GTP

DiaRho

GTP

Cyclin D1

RacGTP

P

Fig. 2. Molecular mechanisms for integration of adhesive signals. Activation of Rac by both cadherins (dark blue) and integrins (light green) upregulatesproliferation in an additive manner through an increase in the expression of cyclin D1 (orange arrows). Cyclic cell strain induces cadherin-dependent activation ofRac and cyclin D1, but it has not yet been established how sensitive integrin-mediated promotion of proliferation is to cyclic strain. Cadherins and integrinsantagonistically influence the activity of Rho GTPase and thus of Rho (purple arrows). Active Rho has dramatically different effects on cell adhesion depending onthe downstream effector it binds. Rho signaling through Dia produces reorganization of actin in a manner that strengthens cell–cell adhesion, whereas signalingthrough ROCK enhances actin contractility, which in turn promotes cell–cell and cell–matrix adhesion. However, a high level of Rho- and ROCK-mediated actincontractility is antagonistic to cell–cell junctions. Fer kinase is activated by cadherin and integrin adhesions, and activated Fer phosphorylates (P) the actin-organizing protein cortactin (blue arrows). Reorganization of the actin cytoskeleton by cortactin can enhance either cell–cell adhesion or migration. Shear stressacross endothelial cells leads to inflammation (green arrows). Shear stress induces the assembly of a VEGFR–VE-cadherin–Shc complex (VEGFR in purple) andthe phosphorylation of Shc, which then leads to activation of ERK1/2 and, subsequently, to inflammation. Phosphorylated Shc associates with integrins in acadherin dependent manner and activates the NF-B pathway, also resulting in inflammation. Solid lines represent direct interactions or effects, dashed linesindicate an indirect or unknown mechanism. The extracellular matrix is shown beneath the cells in blue and the actin cytoskeleton is represented by the pink lines.

Jour

nal o

f Cel

l Sci

ence

Page 5: Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

integrin engagement and activation of Fak (Wang et al., 2004). Fakthen promotes E-cadherin expression and cell cohesion (Wang etal., 2004), but the mechanism by which this occurs remains unclear.Nonetheless, this is consistent with Fak knockdown studies, whichreport stimulation of EMT and inhibition of cadherin adhesion(Yano et al., 2004). Because Fak is normally associated withintegrin adhesions there is much speculation about its role in cell–cell junctions. Regulation of RhoGTPases is one possiblemechanism of action because Fak can inhibit the activity of Rho,and constitutively active Rho mutants can phenocopy Fak loss-of-function (Playford et al., 2008). Interestingly, Fak has also beenshown to localize to cell–cell contacts, although the significance –if any – of its localization at cell–cell adhesions is unclear (Crawfordet al., 2003; Playford et al., 2008).

The non-receptor tyrosine kinase Fer is also reported to beinvolved in communication between cell–cell and cell–ECMadhesive contacts (Arregui et al., 2000). Fer can be activatedupon engagement of either cadherins or integrins (El Sayegh etal., 2005; Sangrar et al., 2007). The association of Fer with theactin-organizing protein cortactin might help coordinate cellularresponse to various adhesive inputs. Cortactin is required for cellspreading on fibronectin, and the activation of cortactin by Ferpromotes cell motility (Illes et al., 2006; Sangrar et al., 2007).Phosphorylation of cortactin by Src and/or Fer downstream of E-cadherin ligation enhances cell–cell adhesion (El Sayegh et al.,2005; Ren et al., 2009). This suggests that Fer signaling feedsback to cell–cell and cell–matrix adhesions by promotingcortactin-dependent reorganization of actin (Fig. 2).

Several growth factor receptor tyrosine kinases are reported toregulate cadherin and integrin adhesive functions. For instance,vascular endothelial growth factor (VEGF) signalling throughthe VEGF receptor 2 (VEGFR2, also known as KDR) both

increases integrin-dependent migration and decreases stability ofvascular endothelial (VE)–cadherin adhesions (Carmeliet et al.,1999; Byzova et al., 2000). Receptor tyrosine kinases can alsoenable lateral molecular associations between different types ofadhesion. IGF1R forms a ternary complex with E-cadherin andv-integrins at cell–cell contacts. Binding of the ligand IGF1causes the relocalization of v-integrin to focal contacts and anincrease in cell migration (Canonici et al., 2008). Thus, IGF1Rmight sequester integrins at cell–cell contacts and inhibitmigration in the absence of growth factor signaling.

PhosphatasesGiven the central importance of kinases to many adhesion-dependent cell signaling pathways, it is crucial to also consider therole of phosphatases in the regulation of adhesive networks. Thereare a number of protein tyrosine phosphatases (PTPs) that associatewith cell–cell and cell–ECM adhesions – such as PTP1B, whichlocalizes to the cytoplasmic tail of cadherins and also to focaladhesions (Stoker, 2005; Burridge et al., 2006; Sallee et al., 2006).The catalytic activity of PTP1B is required for dephosphorylationof -catenin and association of N-cadherin with the actincytoskeleton, both of which are crucial for the stability of cell–celljunctions (Sallee et al., 2006). PTP1B is also involved in signalingfrom integrin adhesions, probably through dephosphorylation ofthe inhibitory tyrosine residue of Src, which leads to activationof Fak (Arregui et al., 1998). Outgrowth of neurites depends onboth cell–cell and cell–matrix interactions, and PTP1B has beenshown to be required for this process, probably also throughregulation of Src activity (Pathre et al., 2001). PTP is anothermember of the PTP family that is necessary for maintaining theintegrity of cell–cell adhesions. In addition to its role in stabilizingcell–cell junctions through dephosphorylation of junctionalcomponents, PTP might also recruit regulatory proteins to sitesof cell–cell adhesion (Sallee et al., 2006). Expression of PTP inkidney epithelial cells is dependent on a complex between 31-integrin and tetraspanin that associates with E-cadherin on thelateral membranes of cells. An interesting feature of this lateralcomplex coupling is that a specific subset of unligated integrinsappears to associate with and promote stability of cell–celladhesions, and that these are distinct from the subset of integrinsinvolved in matrix adhesion (Chattopadhyay et al., 2003).

Scaffolding and adaptor proteinsThe consequences of cadherin and integrin engagement can varygreatly depending upon the specific intracellular environment at thesite of signal initiation. Scaffolding and adaptor proteins are crucialelements of adhesion-dependent signaling cascades. They facilitatethe localization of key downstream effectors, thereby increasing theprobability of interactions between activated signaling components.Receptor for activated C kinase-1 (RACK1, also known as GNB2L1)is a scaffolding protein that binds the cytoplasmic tails of integrinsand interacts with intracellular signaling components such as proteinkinase C (PKC) (Besson et al., 2002). RACK1 is a crucial componentof the E-cadherin and 31-integrin–tetraspanin lateral signalingcomplex described above (Chattopadhyay et al., 2003). Binding toPTP might recruit RACK1 to cell–cell adhesions, serving to localizeactivated PKC or other effectors (Mourton et al., 2001). PKC hasnumerous roles in promoting cell–cell and cell–matrix adhesions(Larsson, 2006). Localization of active PKC by RACK1 might bekey to specifying the types of adhesion that are affected by PKCsignaling.

1187Adhesive crosstalk

Box 2. Rap1 is poised between cadherins andintegrinsRac, Rho and Cdc42 are not the only GTPases functioning in thesignaling network that connects integrins and cadherins. Rap1, amember of the RasGTPase family has also been implicated in thetransmission of signals between cell–cell and cell–matrixadhesions (Retta et al., 2006). It is known to activate integrins invarious cell types, demonstrating a role for Rap1 in inside-outintegrin signaling (Retta et al., 2006). Rap1 was also shown to beimportant for the formation and maintenance of cadherinadhesions (Watanabe et al., 2009). These findings place Rap1upstream of both adhesion types, but more recent work hasdemonstrated the regulation of Rap1 activity by E-cadherinadhesion (Balzac et al., 2005). Disruption of E-cadherin adhesionsresults in a dramatic increase in Rap1 activity, which can then bedownregulated by re-forming cell–cell junctions. Although thisfinding suggests that Rap1 functions both upstream anddownstream of cadherins, the same is not true for integrins. Rap1activity is not dependent on the substrate onto which the cells areplated, indicating that Rap1 is not regulated by integrin adhesions.Dissolution of cell–cell junctions does result in an increase in focaladhesions, and this is abrogated by inhibition of Rap1 activity. Thisplaces Rap1 neatly between cadherin and integrin adhesions. It isnot yet clear how Rap1 activity is affected by endogenousdisruption of cadherin adhesion, such as through increasedactomyosin-mediated tension. Additional data suggest thatinternalization and endocytic trafficking of cadherins is required forRap1 activation, although the precise mechanism of Rap1regulation by cadherins is unknown.

Jour

nal o

f Cel

l Sci

ence

Page 6: Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

In general, adaptor proteins link signaling components butlack intrinsic enzymatic or kinase activities. Motifs such as thephosphotyrosine-binding Src homology 2 (SH2) domain allowadaptors to bring signaling proteins together and propagatesignaling cascades initiated by a diverse array of stimuli. TheSH2 domain-containing (Shc) adaptor protein forms a complexwith growth factor receptor-bound protein 2 (GRB2) and the Sonof Sevenless (SOS) family of guanine nucleotide exchange factorsto activate Ras and, subsequently, mitogen-activated protein(MAP) kinases, the pathway of which is activated through manyinputs including integrin signaling and mechanical force(Ravichandran, 2001). Shc has recently been shown to associatewith both cell–cell and cell–matrix adhesions in response to fluidflow across vascular endothelial cells (Liu et al., 2008).Phosphorylated Shc is recruited to cell–cell junctions at the onsetof fluid flow, particularly in areas of tissue where fluid-flowshear stresses are high and result in inflammation. Previous workdemonstrated the assembly of a VEGFR2–VE-cadherin complexin response to shear stress and it is now apparent that Shc interactswith components of this complex (Shay-Salit et al., 2002; Liu etal., 2008). Shc is also recruited to integrin adhesions in responseto fluid flow, a process that requires the presence of VE-cadherin.Phosphorylation of the MAPK family members ERK1/2 andactivation of NF-B are increased by fluid flow in a Shc-dependent manner. Interestingly the activation of NF-B, but notthe activation of ERK1/2, is dependent on ECM composition(Liu et al., 2008). These data suggest that Shc is a central mediatorof fluid flow-induced signaling events that include cadherin-dependent activation of signaling at cell–matrix junctions (Fig. 2).

There are several other classes of protein that can act as adaptorseven though they are not traditionally thought of as such. Oneexample is the catenin family of proteins, of which some areinvolved in both cell–cell adhesion and cell signaling. Cateninsare crucial for connecting cadherins to the cytoskeleton, althoughnot necessarily through a direct physical link (Nelson, 2008).Catenins are required for the initiation of many signaling pathwaysdownstream of cadherin adhesion, including those that regulateRhoGTPase activities, actomyosin organization and contractility,and stability of cell–cell junctions (Perez-Moreno and Fuchs, 2006).Plakoglobin (Pkg, also known as JUP or g-catenin), associates withboth desmosomes and adherens junctions where it helps maintaintissue integrity through interactions with -catenin or desmoplakin(Zhurinsky et al., 2000; Acehan et al., 2008). By serving as a linkbetween cadherin and either -catenin or desmoplakin, Pkgconnects cadherins to actin or intermediate filament networks,respectively. Pkg is reported to suppress motility of cell sheets andsingle keratinocytes on collagen (Yin et al., 2005). Although themechanism for regulation of cell motility by Pkg is not clear, thereis some evidence that Pkg works through long-range mechanismsto promote fibronectin expression and inhibit Src function (Yin etal., 2005; Todorovic et al., 2010). Pkg is a known target of Srckinase activity, thus it is possible that the presence of Pkg at cell–cell junctions sequesters Src away from cell–matrix adhesionswhere it typically promotes cell migration (Webb et al., 2004; Leeet al., 2010).

CytoskeletonOne consequence of the signalling events and mechanical linkagesthat are initiated by both cell–cell and cell–matrix adhesions is theassembly and reorganization of cytoskeletal networks. Thusthe regulation of cytoskeletal dynamics might be considered a

consequence of convergent signaling. In turn, the cytoskeletonmediates both short-range and long-range physical interactionsbetween adhesions throughout the cell. Linkage to the cytoskeletonprovides structural integrity to adhesions and allows for cellularmovement, maintenance of cell and tissue shape, and remodelingof the extracellular environment. Moreover, by coupling tomolecular motors the cytoskeleton can exert forces that are thenapplied externally to the ECM and neighbouring cells.

Actin filaments provide integral support to the cell by linkingadhesive contacts on the cell surface to the interior. Actin is tetheredby macromolecular complexes at points of integrin contact withECM, and this association can be visualized in vitro on planarsubstrates as focal adhesions. Cadherin adhesions direct theassembly of actin filaments near the cell cortex where they providestrength to lateral contacts between adjacent cells. We refer thereader to a recent review article (Maruthamuthu et al., 2010),which details and summarizes a plethora of reported connectionsbetween cell adhesion molecules and the actin cytoskeleton. Theoverall emerging picture is that – by acting as a force-bearingscaffold between sites of integrin and cadherin adhesion – the actincytoskeleton provides a direct physical basis for adhesive networkinteractions.

Microtubule dynamic instability provides another mechanismfor integrating the cytoskeleton with signaling pathways that areinitiated by both cell–ECM and cell–cell adhesions. Microtubulesare in a state of constant flux through processes of assembly anddisassembly. Extension of microtubule plus ends into focal contactspromotes the dissolution of these adhesions (Ezratty et al., 2005).Association of microtubules with focal contacts promotesphosphorylation of kinases, including Fak, within the focal adhesioncomplex, inducing destabilization of protein–protein associationsand resulting in the release of actin filaments from these contacts.Moreover, cadherins are reported to affect microtubule dynamics.In highly migratory and undifferentiated cells, the minus ends ofmicrotubules are typically anchored in centrosomes. In terminallydifferentiated cells such as polarized epithelia, however, the minusends of microtubules are instead stabilized by association withcadherin adhesions (Chausovsky et al., 2000; Meng et al., 2008).Likewise, plus ends of microtubules in these cells are stabilized bya linkage to the basal cortex through integrin adhesions (Hotta etal., 2010). Thus, microtubules that are capable of regulating cell–ECM adhesions are also anchored and regulated by cadherins,providing another potential cytoskeleton-based mechanism foradhesive networking.

Mitotic spindle orientation is a major determinant of the axisand symmetry of cell division (Bringmann and Hyman, 2005),which is established by the positioning of distally locatedmicrotubule minus-end anchors, the centrosomes (Heald et al.,1997). In epithelial cells, centrosomes are localized to adherensjunctions during mitosis allowing symmetrical cell division withinthe plane of the tissue (Lu et al., 2001; den Elzen et al., 2009). Inother cell types – such as, for example, neural stem cells – cadherinand integrin adhesions are hypothesized to compete for microtubuleassociation with centrosomes (Loulier et al., 2009). Because of thiscompetition, mitotic spindle orientation and cell division occur onan oblique axis. The result is an asymmetric cell division thatleaves the neural stem cell in its niche while creating a daughtercell that moves into more differentiated regions of the centralnervous system (Kosodo et al., 2004). Although the precisemechanisms remain unclear, coordinated positioning of the spindleby the adhesion receptor network appears to have broad significance

1188 Journal of Cell Science 124 (8)

Jour

nal o

f Cel

l Sci

ence

Page 7: Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

to adult stem cell niche maintenance (Marthiens et al., 2010),organ development (Baena-Lopez et al., 2005), and early embryonicmorphogenesis (Gong et al., 2004).

Intermediate filaments also associate directly with both integrinand cadherin adhesions. In contrast to actin filaments, intermediatefilaments exhibit high tensile strength, extensibility, elasticity andflexibility (Fudge et al., 2003). Nonetheless, intermediate filamentsare highly dynamic and capable of depolymerization andpolymerization anywhere along an existing filament (Godsel et al.,2008; Colakoglu and Brown, 2009). Although most studies ofintermediate filaments have focused on their association withhighly stable adhesive junctions, such as desmosomes andhemidesmosomes, they are also found in association with moredynamic adhesions that involve classical cadherins (Kowalczyk etal., 1998; Leonard et al., 2008) and non-hemidesmosomal integrins(Tsuruta and Jones, 2003; Bhattacharya et al., 2009). Intermediatefilaments lack filament polarity, and the distinct ‘hubs’ of filamentorganization and polymerization (Godsel et al., 2008) that arecharacteristic of the actin and microtubule cytoskeletons.Intermediate filament networks have yet to be implicated directlyin functionally connecting different adhesions; however, their rolein anchoring and stabilizing adhesive junctions suggests that theyare important, particularly in cases where adhesive networks aresubject to mechanical forces.

Different types of cytoskeletal network are further integratedthrough direct crosslinking by cytoskeletal scaffold proteins suchas plectins, which contain binding sites for actin, microtubules andintermediate filaments (Sonnenberg and Liem, 2007). Moreover,motor proteins, such as myosin, kinesin and dynein, can be thoughtof as mobile crosslinkers that generate forces within and betweencytoskeletal networks (Chang and Goldman, 2004). As aconsequence of these varied molecular crosslinks, cadherin andintegrin adhesions are able to form and maintain physicalconnections that are not necessarily limited to a single cytoskeletalnetwork.

Adhesive networks in mechanotransductionand multicellular processesAs signaling through adhesive networks proceeds, the impact oncellular functions is substantial. Many of the pathways discussedabove were elucidated using immortalized tumorigenic cell lines,so the functional implications for normal tissues are notimmediately apparent. However, other studies that focused on avariety of tissues, developmental systems and primary cells haveprovided considerable insight into the physiological roles ofadhesive interactions. Nearly all major cellular functions arereported to be influenced by a combination of integrin and cadherinsignaling events, including proliferation, migration, differentiation(Box 3) and apoptosis (Fouquet et al., 2004; Kang et al., 2007).Moreover, the coordinated initiation, strengthening and dissolutionof cell–cell and cell–matrix adhesions are crucial for morphogenesisand, thus, the directing of both form and function.

MigrationIn cancer metastasis and EMT, integrin and cadherin adhesionsdisplay an antagonistic relationship that determines whether cellsmaintain associations with their neighbors or uncouple and initiatemigration (Avizienyte et al., 2002; Guarino, 2007). For example,in ovarian carcinoma cells, downregulation of E-cadherin throughmatrix metalloproteinase (MMP)-mediated proteolysis is initiatedby integrin ligation to collagen (Symowicz et al., 2007). This

decreased cadherin-mediated cohesion allows the cells to disperseand migrate independently.

Although this scenario might best reflect the behavior ofmetastatic cancer cells and other instances of EMT, it is importantto consider that normal, intact tissues also undergo migratoryprocesses. In collective cell migration, substrate traction is finelybalanced with cell cohesion to maintain organization of themigratory tissue (Fig. 3A). In fact, cadherin adhesions in normaltissues provide instructive signals that regulate the polarity of cellsand, as a result, the direction of migration. As cells come in contactwith one another, cadherin adhesions are formed, Cdc42 signalingis activated, and the centrosome reorients anteriorly relative to thecell nucleus (Desai et al., 2009; Dupin et al., 2009), indicating thata broad repolarization has occurred throughout the cell. Protrusiveactivity is suppressed locally near cadherin adhesions, and increasedon opposing sides of the cell at sites of integrin–ECM adhesion(Desai et al., 2009; Borghi et al., 2010). Even highly migratorytissues, such as some cranial neural crest, maintain cell–cell

1189Adhesive crosstalk

Box 3. Adhesive interactions in differentiation andmorphogenesis

Cell differentiationDifferentiation of cells is influenced by cell adhesion receptors,which integrate several different adhesive inputs, including ECMcomposition, cell density and cell shape (McBeath et al., 2004;Messina et al., 2005; Engler et al., 2006; Rozario and DeSimone,2010). Actomyosin-generated tension is applied through integrinadhesions and provides an instructive signal for the differentiationof mesenchymal stem cells into myoblasts (Engler et al., 2006;Gao et al., 2010). Integrin-mediated activation of Rac impactscell–cell adhesions through increasing N-cadherin expression, aprocess necessary for myogenesis (Gao et al., 2010). Inmyogenesis, cell–matrix adhesion signaling enhances cell–celladhesions, but in some other cell types cadherins and integrinsantagonize each other to carry out a differentiation program. Forexample, in terminally differentiating keratinocytes, cadherins arerequired for tissue stratification, expression of keratinocyte-specific genes and downregulation of integrins – all crucial stepsin the keratinocyte differentiation program (Hodivala and Watt,1994; Watt, 2002). These types of coordinated interactionbetween cadherins and integrins alter cell signaling, and changetissue architecture to promote the differentiated phenotype.

Branching morphogenesisBranching morphogenesis is a crucial developmental processand responsible for the formation and functional organization oflung, kidney and most glandular tissues. During branchingmorphogenesis, fibronectin engagement by integrins results inlocalized decrease of cadherin expression at sites of cleftformation (Sakai et al., 2003). This is an example of long-rangeinput–output interactions (Fig. 1A), by which – in this caseintegrin – signaling increases the expression of Btbd7. HowBTBD7 operates is not known, but its presence leads to changesin gene expression that include suppression of E-cadherin andinduction of the cell-scattering snail homolog 2 (Snai2)gene(Onodera et al., 2010). Although localized suppression of E-cadherin expression promotes the cleft formation that precedesnew branching, E-cadherin is more broadly required throughoutthe tissue to support columnar morphology, bud outgrowth andlumenal structure (Walker et al., 2008). For branchingmorphogenesis, localized downregulation of cadherins byintegrin–matrix engagement is a way of designating distinct areasof tissue to undergo morphogenetic growth.

Jour

nal o

f Cel

l Sci

ence

Page 8: Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

adhesions while they move, although these adhesions are dynamicand cell contacts transient. N-cadherin adhesions in neural crestcells are required for the subcellular localization of activatedRhoGTPases in response to chemoattractants. Without N-cadherinthe cells fail to migrate directionally or collectively (Theveneau etal., 2010). Expression of N-cadherin at the cell surface is regulatedlocally through signaling pathways that are initiated by 1- and3-integrins (Monier-Gavelle and Duband, 1997). In border cellsof Drosophila melanogaster, asymmetric protrusions initiated bylocalized Rac activation in a single border cell can alter migratorydirection of an entire cluster of border cells (Wang et al., 2010).Cohesion of border cells is stabilized by both -integrin and Racsignaling through the MAPK Jun N-terminal kinase (JNK), andthese signaling proteins are required for normal collective cellmigration (Llense and Martin-Blanco, 2008; Wang et al., 2010).

Migration of whole tissues is a recurring feature of developmentand morphogenesis, and wound healing. Collectively migratingtissues use a mechanism of distributed traction, whereby bothleading edge cells and those that follow polarize and migrate in adirectional manner (Davidson et al., 2002; Farooqui and Fenteany,2005). Substrate traction forces and intercellular tissue tension aredeveloped as an intact tissue translocates. Trepat and co-workersreported that, although leading cells generate the highest substratetraction, intercellular tension increases progressively as a functionof distance from the leading edge (Trepat et al., 2009). Moreover,tension on cadherin adhesions recruits scaffolding proteins, suchas vinculin and -catenin, which mediate cytoskeletal reinforcementof the adhesions and the resultant strain-stiffening response (Chuet al., 2004; le Duc et al., 2010; Yonemura et al., 2010). However,an important question remains when tying these observations

together: does tension on cadherin adhesions have ramificationsfor protrusive activity and polarity of the cell? We speculate that,because cadherins are responsive to mechanical force, changes inthe physical linkage of cadherins to cytoskeletal networks and theassociation with scaffolding proteins have an important role inthe polarization of migratory cells and tissues.

Early embryonic morphogenesisSeveral examples of adhesive networks can be illustrated by usingearly-embryonic models of tissue morphogenesis. Duringconvergent extension, mesodermal cells mediolaterally intercalateto form the notochord and drive axial elongation of the embryo(Fig. 3B). The cells are not able to complete this process in theabsence of fibronectin, when 51-integrin is inhibited, or whencadherin adhesion is altered (Marsden and DeSimone, 2003;Davidson et al., 2006). Moreover, adhesion and signaling of 51-integrin can modulate cadherin adhesion, which is required for cellintercalation and cell-sorting behaviors (Marsden and DeSimone,2003), although the mechanism of signaling between theseadhesions remains unclear. Communication between adhesionreceptors in this system is also bidirectional. Tension on cadherinadhesions induces integrin-dependent assembly of fibronectin fibrils(Fig. 3C) (Dzamba et al., 2009). Fibronectin fibril assembly is, inturn, required for normal morphogenetic movements in earlyembryogenesis, such as epiboly and mesendodermal migration(Rozario et al., 2009). Activation of the planar cell polarity pathway(PCP) – a non-canonical Wnt signaling cascade – is required forconvergence and extension movements, and also promotes theassembly of the fibronectin matrix by regulating cadherin adhesionand tissue tension (Dzamba et al., 2009). Thus, PCP signaling is

1190 Journal of Cell Science 124 (8)

Fluid-flow shear stress Cyclic strain

Migration

Hydrostaticcompression

D Vascular endothelium

B Convergent extension

A Collective migration

Axial extension

C In vivo matrix assembly

Fig. 3. Mechanical inputs modulate adhesive networks incomplex tissue systems. (A)Collective migration involvestraction forces through integrin–matrix adhesions to drive thetissue forward (green arrows). Extracellular matrix substratumis in purple. The tissue advances against intercellular tissuetension (pink arrows) that is mediated by cadherins (pinkrectangles). This intercellular tension is anisotropicallydistributed in the tissue and is greater within the tissue than atthe leading edge. (B)Convergence and extension movementsin gastrulation involve coordinated regulation of differentadhesion types. Bidirectional protrusive activity (greenarrows) mediated by PCP signaling and integrin–matrixadhesions enables sliding of cells past each other, which is acadherin-dependent process (cadherin adhesions are shown aspink rectangles). Intercalation behavior creates force inanterior and posterior directions, thereby driving axialelongation. (C)In vivo, assembly of matrix (purple lines) canbe regulated by force transduced through cadherin adhesions(pink rectangles). In multicellular tissues, cells pull on oneanother (pink arrows) through cadherin adhesions to increaselocal mechanical tension. It has been proposed that integrinstranslocate from sites of cell–cell contact across the freesurface of the cell (green arrows) and promote matrixassembly. (D)Endothelial cells in vivo are simultaneouslyexposed to fluid-flow shear stress, cyclic strain andhydrostatic compression (large open arrows), eachrepresenting a mechanical stress that applies force to cadherinand integrin adhesions (filled arrows, color matched to theapplicable force). Mechanical forces on cadherins andintegrins initiate input–output and convergent type signalingacross the adhesive network to regulate cell morphology andphysiological responses such as cell proliferation.

Jour

nal o

f Cel

l Sci

ence

Page 9: Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

probably an important signaling pathway that links cadherin andintegrin functions in the early embryo.

Endothelial cell biologyVascular endothelial cells comprise the inner layer of blood vesselsand are exposed to three major types of mechanical stress: shearstress, cyclic strain and hydrostatic pressure (Fig. 3D). These cellsare in tight cohesion with one another and with the underlyingmatrix, forming a semipermeable barrier against the pressurizedbloodstream. Mechanical stress on this tissue is sensed andtransduced, at least in part, by adhesion complexes. Shear stressinduces alignment and elongation of cells in the direction of fluidflow, as well as expression of genes that result in an inflammatoryresponse. VE-cadherin, platelet endothelial cell adhesion molecule(PECAM) and VEGFR2 form a complex in response to flow, andthis complex signals through Shc to activate the integrin responseto shear (Fig. 2) (Tzima et al., 2005; Liu et al., 2008). Cyclic straininduces proliferation in endothelial cells through a VE-cadherin–Rac1-mediated pathway (Liu et al., 2007). Integrin signaling hasbeen demonstrated to increase cyclin D1 in endothelial cells topromote proliferation (Schwartz and Assoian, 2001), and cell straininduces proliferation in some cell types through integrin signaling(Wilson et al., 1995). It remains to be determined, however, whetherthe proliferative effects of cyclic strain are mediated entirely byVE-cadherin or whether strain induction of endothelial proliferationalso requires integrin-mediated cyclin D1 expression.

Proliferation can also be stimulated by signaling through v-integrins in endothelial cells under hydrostatic pressure (Fig. 2)(Schwartz et al., 1999). However, the signal to proliferate isnegatively balanced by the presence of VE-cadherin-mediated cellcontacts (Ohashi et al., 2007), which are stabilized by physiologicalhydrostatic pressures (Muller-Marschhausen et al., 2008). Thepoint at which VE-cadherin and v-integrin signals converge toregulate proliferation induced through hydrostatic pressure isunknown. Furthermore, it is not clear how VE-cadherins negativelyregulate proliferation in this context yet promote proliferation thatis induced by cyclic strain. Clearly, different types of stress oncadherin adhesions stimulate unique signaling pathways thatdifferentially affect integrin signaling. Endothelial cells in vivo areexposed to multiple stresses simultaneously. The mechanism bywhich cells integrate these disparate inputs and orchestrate a cellularresponse is an important question for future research.

ConclusionsIn this commentary we have presented an overview of emergingmechanisms through which cadherin- and integrin-dependentadhesion and signaling functions intersect and interact. Althoughcell–cell and cell–ECM adhesive complexes have variablecompositions and mediate interactions at often topologically distinctpositions in the cell, they share many structural similarities andsignaling functions. Cadherins and integrins each coordinateresponses to changes in the adhesive state through altered physicallinkages with the cytoskeleton and by participating in bidirectionalcell signaling events. Rather than thinking of cell–cell and cell–matrix adhesions in individual cells and tissues as operating largelyin isolation, we argue that these complexes are part of a largeradhesive network wherein multiple types of cell adhesionnecessarily interact. Much future effort in this area will continueto focus on elucidating the remaining signaling molecules,scaffolding and adapter proteins that comprise adhesive networks.However, the larger challenge is to understand how cell–cell and

cell–ECM adhesion and signaling inputs are integrated to affectthe responses of cells within contextually appropriate and dynamicmicroenvironments, such as those found within embryos, normaltissues and tumors.

We thank our colleagues Bette Dzamba, Tania Rozario and ShuoWei for reading and commenting on the manuscript. The authors weresupported by USPHS grants F32-GM83542 to G.F.W., T32-GM08136to M.A.B., and R01-HD26402/GM094793 to D.W.D. Deposited inPMC for release after 12 months.

ReferencesAcehan, D., Petzold, C., Gumper, I., Sabatini, D. D., Muller, E. J., Cowin, P. and

Stokes, D. L. (2008). Plakoglobin is required for effective intermediate filamentanchorage to desmosomes. J. Invest. Dermatol. 128, 2665-2675.

Arregui, C. O., Balsamo, J. and Lilien, J. (1998). Impaired integrin-mediated adhesionand signaling in fibroblasts expressing a dominant-negative mutant PTP1B. J. Cell Biol.143, 861-873.

Arregui, C., Pathre, P., Lilien, J. and Balsamo, J. (2000). The nonreceptor tyrosinekinase fer mediates cross-talk between N-cadherin and beta1-integrins. J. Cell Biol.149, 1263-1274.

Avizienyte, E., Wyke, A. W., Jones, R. J., McLean, G. W., Westhoff, M. A., Brunton,V. G. and Frame, M. C. (2002). Src-induced de-regulation of E-cadherin in coloncancer cells requires integrin signalling. Nat. Cell Biol. 4, 632-638.

Avizienyte, E., Fincham, V. J., Brunton, V. G. and Frame, M. C. (2004). Src SH3/2domain-mediated peripheral accumulation of Src and phospho-myosin is linked toderegulation of E-cadherin and the epithelial-mesenchymal transition. Mol. Biol. Cell15, 2794-2803.

Baena-Lopez, L. A., Baonza, A. and Garcia-Bellido, A. (2005). The orientation of celldivisions determines the shape of Drosophila organs. Curr. Biol. 15, 1640-1644.

Balzac, F., Avolio, M., Degani, S., Kaverina, I., Torti, M., Silengo, L., Small, J. V. andRetta, S. F. (2005). E-cadherin endocytosis regulates the activity of Rap1: a traffic lightGTPase at the crossroads between cadherin and integrin function. J. Cell Sci. 118, 4765-4783.

Bass, M. D., Morgan, M. R., Roach, K. A., Settleman, J., Goryachev, A. B. andHumphries, M. J. (2008). p190RhoGAP is the convergence point of adhesion signalsfrom alpha 5 beta 1 integrin and syndecan-4. J. Cell Biol. 181, 1013-1026.

Behrens, J., Vakaet, L., Friis, R., Winterhager, E., Van Roy, F., Mareel, M. M. andBirchmeier, W. (1993). Loss of epithelial differentiation and gain of invasivenesscorrelates with tyrosine phosphorylation of the E-cadherin/beta-catenin complex incells transformed with a temperature-sensitive v-SRC gene. J. Cell Biol. 120, 757-766.

Besson, A., Wilson, T. L. and Yong, V. W. (2002). The anchoring protein RACK1 linksprotein kinase Cepsilon to integrin beta chains. Requirements for adhesion and motility.J. Biol. Chem. 277, 22073-22084.

Bhadriraju, K., Yang, M., Alom Ruiz, S., Pirone, D., Tan, J. and Chen, C. S. (2007).Activation of ROCK by RhoA is regulated by cell adhesion, shape, and cytoskeletaltension. Exp. Cell Res. 313, 3616-3623.

Bhattacharya, R., Gonzalez, A. M., Debiase, P. J., Trejo, H. E., Goldman, R. D.,Flitney, F. W. and Jones, J. C. (2009). Recruitment of vimentin to the cell surface bybeta3 integrin and plectin mediates adhesion strength. J. Cell Sci. 122, 1390-1400.

Borghi, N., Lowndes, M., Maruthamuthu, V., Gardel, M. L. and Nelson, W. J. (2010).Regulation of cell motile behavior by crosstalk between cadherin- and integrin-mediatedadhesions. Proc. Natl. Acad. Sci. USA 107, 13324-13329.

Braga, V. M. and Yap, A. S. (2005). The challenges of abundance: epithelial junctionsand small GTPase signalling. Curr. Opin. Cell Biol. 17, 466-474.

Bringmann, H. and Hyman, A. A. (2005). A cytokinesis furrow is positioned by twoconsecutive signals. Nature 436, 731-734.

Burridge, K., Sastry, S. K. and Sallee, J. L. (2006). Regulation of cell adhesion byprotein-tyrosine phosphatases. I. Cell-matrix adhesion. J. Biol. Chem. 281, 15593-15596.

Byzova, T. V., Goldman, C. K., Pampori, N., Thomas, K. A., Bett, A., Shattil, S. J. andPlow, E. F. (2000). A mechanism for modulation of cellular responses to VEGF:activation of the integrins. Mol. Cell 6, 851-860.

Canonici, A., Steelant, W., Rigot, V., Khomitch-Baud, A., Boutaghou-Cherid, H.,Bruyneel, E., Van Roy, F., Garrouste, F., Pommier, G. and Andre, F. (2008). Insulin-like growth factor-I receptor, E-cadherin and alpha v integrin form a dynamic complexunder the control of alpha-catenin. Int. J. Cancer 122, 572-582.

Carmeliet, P., Lampugnani, M. G., Moons, L., Breviario, F., Compernolle, V., Bono,F., Balconi, G., Spagnuolo, R., Oosthuyse, B., Dewerchin, M. et al. (1999). Targeteddeficiency or cytosolic truncation of the VE-cadherin gene in mice impairs VEGF-mediated endothelial survival and angiogenesis. Cell 98, 147-157.

Chang, L. and Goldman, R. D. (2004). Intermediate filaments mediate cytoskeletalcrosstalk. Nat. Rev. Mol. Cell Biol. 5, 601-613.

Chartier, N. T., Laine, M., Gout, S., Pawlak, G., Marie, C. A., Matos, P., Block, M. R.and Jacquier-Sarlin, M. R. (2006). Laminin-5-integrin interaction signals through PI3-kinase and Rac1b to promote assembly of adherens junctions in HT-29 cells. J. CellSci. 119, 31-46.

Chattopadhyay, N., Wang, Z., Ashman, L. K., Brady-Kalnay, S. M. and Kreidberg,J. A. (2003). alpha3beta1 integrin-CD151, a component of the cadherin-catenin complex,regulates PTPmu expression and cell-cell adhesion. J. Cell Biol. 163, 1351-1362.

1191Adhesive crosstalk

Jour

nal o

f Cel

l Sci

ence

Page 10: Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

Chausovsky, A., Bershadsky, A. D. and Borisy, G. G. (2000). Cadherin-mediatedregulation of microtubule dynamics. Nat. Cell Biol. 2, 797-804.

Chu, Y. S., Thomas, W. A., Eder, O., Pincet, F., Perez, E., Thiery, J. P. and Dufour, S.(2004). Force measurements in E-cadherin-mediated cell doublets reveal rapid adhesionstrengthened by actin cytoskeleton remodeling through Rac and Cdc42. J. Cell Biol.167, 1183-1194.

Colakoglu, G. and Brown, A. (2009). Intermediate filaments exchange subunits alongtheir length and elongate by end-to-end annealing. J. Cell Biol. 185, 769-777.

Crawford, B. D., Henry, C. A., Clason, T. A., Becker, A. L. and Hille, M. B. (2003).Activity and distribution of paxillin, focal adhesion kinase, and cadherin indicatecooperative roles during zebrafish morphogenesis. Mol. Biol. Cell 14, 3065-3081.

Davidson, L. A., Hoffstrom, B. G., Keller, R. and DeSimone, D. W. (2002). Mesendodermextension and mantle closure in Xenopus laevis gastrulation: combined roles for integrinalpha(5)beta(1), fibronectin, and tissue geometry. Dev. Biol. 242, 109-129.

Davidson, L. A., Marsden, M., Keller, R. and Desimone, D. W. (2006). Integrinalpha5beta1 and fibronectin regulate polarized cell protrusions required for Xenopusconvergence and extension. Curr. Biol. 16, 833-844.

den Elzen, N., Buttery, C. V., Maddugoda, M. P., Ren, G. and Yap, A. S. (2009).Cadherin adhesion receptors orient the mitotic spindle during symmetric cell divisionin mammalian epithelia. Mol. Biol. Cell 20, 3740-3750.

Desai, R. A., Gao, L., Raghavan, S., Liu, W. F. and Chen, C. S. (2009). Cell polaritytriggered by cell-cell adhesion via E-cadherin. J. Cell Sci. 122, 905-911.

Dupin, I., Camand, E. and Etienne-Manneville, S. (2009). Classical cadherins controlnucleus and centrosome position and cell polarity. J. Cell Biol. 185, 779-786.

Dzamba, B. J., Jakab, K. R., Marsden, M., Schwartz, M. A. and DeSimone, D. W.(2009). Cadherin adhesion, tissue tension, and noncanonical Wnt signaling regulatefibronectin matrix organization. Dev. Cell 16, 421-432.

Ehrlich, J. S., Hansen, M. D. and Nelson, W. J. (2002). Spatio-temporal regulation ofRac1 localization and lamellipodia dynamics during epithelial cell-cell adhesion. Dev.Cell 3, 259-270.

El Sayegh, T. Y., Arora, P. D., Fan, L., Laschinger, C. A., Greer, P. A., McCulloch, C.A. and Kapus, A. (2005). Phosphorylation of N-cadherin-associated cortactin by Ferkinase regulates N-cadherin mobility and intercellular adhesion strength. Mol. Biol. Cell16, 5514-5527.

Engler, A. J., Sen, S., Sweeney, H. L. and Discher, D. E. (2006). Matrix elasticity directsstem cell lineage specification. Cell 126, 677-689.

Etienne-Manneville, S. and Hall, A. (2002). Rho GTPases in cell biology. Nature 420,629-635.

Ezratty, E. J., Partridge, M. A. and Gundersen, G. G. (2005). Microtubule-inducedfocal adhesion disassembly is mediated by dynamin and focal adhesion kinase. Nat.Cell Biol. 7, 581-590.

Farooqui, R. and Fenteany, G. (2005). Multiple rows of cells behind an epithelial woundedge extend cryptic lamellipodia to collectively drive cell-sheet movement. J. Cell Sci.118, 51-63.

Fouquet, S., Lugo-Martinez, V. H., Faussat, A. M., Renaud, F., Cardot, P., Chambaz,J., Pincon-Raymond, M. and Thenet, S. (2004). Early loss of E-cadherin from cell-cell contacts is involved in the onset of Anoikis in enterocytes. J. Biol. Chem. 279,43061-43069.

Fournier, A. K., Campbell, L. E., Castagnino, P., Liu, W. F., Chung, B. M., Weaver,V. M., Chen, C. S. and Assoian, R. K. (2008). Rac-dependent cyclin D1 geneexpression regulated by cadherin- and integrin-mediated adhesion. J. Cell Sci. 121, 226-233.

Fudge, D. S., Gardner, K. H., Forsyth, V. T., Riekel, C. and Gosline, J. M. (2003). Themechanical properties of hydrated intermediate filaments: insights from hagfish slimethreads. Biophys. J. 85, 2015-2027.

Gao, L., McBeath, R. and Chen, C. S. (2010). Stem cell shape regulates a chondrogenicversus myogenic fate through Rac1 and N-cadherin. Stem Cells 28, 564-572.

Giannone, G. and Sheetz, M. P. (2006). Substrate rigidity and force define form throughtyrosine phosphatase and kinase pathways. Trends Cell Biol. 16, 213-223.

Godsel, L. M., Hobbs, R. P. and Green, K. J. (2008). Intermediate filament assembly:dynamics to disease. Trends Cell Biol. 18, 28-37.

Gong, Y., Mo, C. and Fraser, S. E. (2004). Planar cell polarity signalling controls celldivision orientation during zebrafish gastrulation. Nature 430, 689-693.

Guarino, M. (2007). Epithelial-mesenchymal transition and tumour invasion. Int. J.Biochem. Cell Biol. 39, 2153-2160.

Hall, A. (1998). Rho GTPases and the actin cytoskeleton. Science 279, 509-514.Heald, R., Tournebize, R., Habermann, A., Karsenti, E. and Hyman, A. (1997).

Spindle assembly in Xenopus egg extracts: respective roles of centrosomes andmicrotubule self-organization. J. Cell Biol. 138, 615-628.

Hodivala, K. J. and Watt, F. M. (1994). Evidence that cadherins play a role in thedownregulation of integrin expression that occurs during keratinocyte terminaldifferentiation. J. Cell Biol. 124, 589-600.

Hotta, A., Kawakatsu, T., Nakatani, T., Sato, T., Matsui, C., Sukezane, T., Akagi, T.,Hamaji, T., Grigoriev, I., Akhmanova, A. et al. (2010). Laminin-based cell adhesionanchors microtubule plus ends to the epithelial cell basal cortex through LL5alpha/beta.J. Cell Biol. 189, 901-917.

Huveneers, S. and Danen, E. H. (2009). Adhesion signaling-crosstalk between integrins,Src and Rho. J. Cell Sci. 122, 1059-1069.

Illes, A., Enyedi, B., Tamas, P., Balazs, A., Bogel, G., Melinda, Lukacs and Buday, L.(2006). Cortactin is required for integrin-mediated cell spreading. Immunol. Lett. 104,124-130.

Kang, H. G., Jenabi, J. M., Zhang, J., Keshelava, N., Shimada, H., May, W. A., Ng,T., Reynolds, C. P., Triche, T. J. and Sorensen, P. H. (2007). E-cadherin cell-cell

adhesion in ewing tumor cells mediates suppression of anoikis through activation of theErbB4 tyrosine kinase. Cancer Res. 67, 3094-3105.

Kosodo, Y., Roper, K., Haubensak, W., Marzesco, A. M., Corbeil, D. and Huttner, W.B. (2004). Asymmetric distribution of the apical plasma membrane during neurogenicdivisions of mammalian neuroepithelial cells. EMBO J. 23, 2314-2324.

Kowalczyk, A. P., Navarro, P., Dejana, E., Bornslaeger, E. A., Green, K. J., Kopp, D.S. and Borgwardt, J. E. (1998). VE-cadherin and desmoplakin are assembled intodermal microvascular endothelial intercellular junctions: a pivotal role for plakoglobinin the recruitment of desmoplakin to intercellular junctions. J. Cell Sci. 111, 3045-3057.

Larsson, C. (2006). Protein kinase C and the regulation of the actin cytoskeleton. Cell.Signal. 18, 276-284.

le Duc, Q., Shi, Q., Blonk, I., Sonnenberg, A., Wang, N., Leckband, D. and de Rooij,J. (2010). Vinculin potentiates E-cadherin mechanosensing and is recruited to actin-anchored sites within adherens junctions in a myosin II-dependent manner. J. Cell Biol.189, 1107-1115.

Lee, H. H., Tien, S. C., Jou, T. S., Chang, Y. C., Jhong, J. G. and Chang, Z. F. (2010).Src-dependent phosphorylation of ROCK participates in regulation of focal adhesiondynamics. J. Cell Sci. 123, 3368-3377.

Leonard, M., Chan, Y. and Menko, A. S. (2008). Identification of a novel intermediatefilament-linked N-cadherin/gamma-catenin complex involved in the establishment ofthe cytoarchitecture of differentiated lens fiber cells. Dev. Biol. 319, 298-308.

Liu, W. F., Nelson, C. M., Tan, J. L. and Chen, C. S. (2007). Cadherins, RhoA, and Rac1are differentially required for stretch-mediated proliferation in endothelial versus smoothmuscle cells. Circ. Res. 101, e44-e52.

Liu, Y., Sweet, D. T., Irani-Tehrani, M., Maeda, N. and Tzima, E. (2008). Shccoordinates signals from intercellular junctions and integrins to regulate flow-inducedinflammation. J. Cell Biol. 182, 185-196.

Liu, Z., Tan, J. L., Cohen, D. M., Yang, M. T., Sniadecki, N. J., Ruiz, S. A., Nelson,C. M. and Chen, C. S. (2010). Mechanical tugging force regulates the size of cell-celljunctions. Proc. Natl. Acad. Sci. USA 107, 9944-9949.

Llense, F. and Martin-Blanco, E. (2008). JNK signaling controls border cell clusterintegrity and collective cell migration. Curr. Biol. 18, 538-544.

Loulier, K., Lathia, J. D., Marthiens, V., Relucio, J., Mughal, M. R., Tang, S. C.,Coksaygan, T., Hall, P. E., Chigurupati, S., Patton, B. et al. (2009). beta1 integrinmaintains integrity of the embryonic neocortical stem cell niche. PloS Biol. 7, e1000176.

Lu, B., Roegiers, F., Jan, L. Y. and Jan, Y. N. (2001). Adherens junctions inhibitasymmetric division in the Drosophila epithelium. Nature 409, 522-525.

Marsden, M. and DeSimone, D. W. (2003). Integrin-ECM interactions regulate cadherin-dependent cell adhesion and are required for convergent extension in Xenopus. Curr.Biol. 13, 1182-1191.

Marthiens, V., Kazanis, I., Moss, L., Long, K. and Ffrench-Constant, C. (2010).Adhesion molecules in the stem cell niche-more than just staying in shape? J. Cell Sci.123, 1613-1622.

Martinez-Rico, C., Pincet, F., Thiery, J. P. and Dufour, S. (2010). Integrins stimulateE-cadherin-mediated intercellular adhesion by regulating Src-kinase activation andactomyosin contractility. J. Cell Sci. 123, 712-722.

Maruthamuthu, V., Aratyn-Schaus, Y. and Gardel, M. L. (2010). Conserved F-actindynamics and force transmission at cell adhesions. Curr. Opin. Cell Biol. 22, 583-588.

McBeath, R., Pirone, D. M., Nelson, C. M., Bhadriraju, K. and Chen, C. S. (2004).Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev.Cell 6, 483-495.

McLachlan, R. W., Kraemer, A., Helwani, F. M., Kovacs, E. M. and Yap, A. S. (2007).E-cadherin adhesion activates c-Src signaling at cell-cell contacts. Mol. Biol. Cell 18,3214-3223.

Meng, W., Mushika, Y., Ichii, T. and Takeichi, M. (2008). Anchorage of microtubuleminus ends to adherens junctions regulates epithelial cell-cell contacts. Cell 135, 948-959.

Messina, G., Blasi, C., La Rocca, S. A., Pompili, M., Calconi, A. and Grossi, M. (2005).p27Kip1 acts downstream of N-cadherin-mediated cell adhesion to promote myogenesisbeyond cell cycle regulation. Mol. Biol. Cell 16, 1469-1480.

Monier-Gavelle, F. and Duband, J. L. (1997). Cross talk between adhesion molecules:control of N-cadherin activity by intracellular signals elicited by beta1 and beta3integrins in migrating neural crest cells. J. Cell Biol. 137, 1663-1681.

Mourton, T., Hellberg, C. B., Burden-Gulley, S. M., Hinman, J., Rhee, A. and Brady-Kalnay, S. M. (2001). The PTPmu protein-tyrosine phosphatase binds and recruits thescaffolding protein RACK1 to cell-cell contacts. J. Biol. Chem. 276, 14896-14901.

Muller-Marschhausen, K., Waschke, J. and Drenckhahn, D. (2008). Physiologicalhydrostatic pressure protects endothelial monolayer integrity. Am. J. Physiol. CellPhysiol. 294, C324-C332.

Nelson, W. J. (2008). Regulation of cell-cell adhesion by the cadherin-catenin complex.Biochem. Soc. Trans. 36, 149-155.

Noren, N. K., Niessen, C. M., Gumbiner, B. M. and Burridge, K. (2001). Cadherinengagement regulates Rho family GTPases. J. Biol. Chem. 276, 33305-33308.

Ohashi, T., Sugaya, Y., Sakamoto, N. and Sato, M. (2007). Hydrostatic pressureinfluences morphology and expression of VE-cadherin of vascular endothelial cells. J.Biomech. 40, 2399-2405.

Onodera, T., Sakai, T., Hsu, J. C., Matsumoto, K., Chiorini, J. A. and Yamada, K. M.(2010). Btbd7 regulates epithelial cell dynamics and branching morphogenesis. Science329, 562-565.

Pathre, P., Arregui, C., Wampler, T., Kue, I., Leung, T. C., Lilien, J. and Balsamo, J.(2001). PTP1B regulates neurite extension mediated by cell-cell and cell-matrix adhesionmolecules. J. Neurosci. Res. 63, 143-150.

Perez-Moreno, M. and Fuchs, E. (2006). Catenins: keeping cells from getting theirsignals crossed. Dev. Cell 11, 601-612.

1192 Journal of Cell Science 124 (8)

Jour

nal o

f Cel

l Sci

ence

Page 11: Integrins and cadherins join forces to form adhesive networks · points of convergent signaling downstream of adhesions as well as upstream modifiers of functions of individual adhesion

Playford, M. P. and Schaller, M. D. (2004). The interplay between Src and integrins innormal and tumor biology. Oncogene 23, 7928-7946.

Playford, M. P., Vadali, K., Cai, X., Burridge, K. and Schaller, M. D. (2008). Focaladhesion kinase regulates cell-cell contact formation in epithelial cells via modulationof Rho. Exp. Cell Res. 314, 3187-3197.

Ravichandran, K. S. (2001). Signaling via Shc family adapter proteins. Oncogene 20,6322-6330.

Ren, G., Helwani, F. M., Verma, S., McLachlan, R. W., Weed, S. A. and Yap, A. S.(2009). Cortactin is a functional target of E-cadherin-activated Src family kinases inMCF7 epithelial monolayers. J. Biol. Chem. 284, 18913-18922.

Ren, X. D., Kiosses, W. B. and Schwartz, M. A. (1999). Regulation of the small GTP-binding protein Rho by cell adhesion and the cytoskeleton. EMBO J. 18, 578-585.

Retta, S. F., Balzac, F. and Avolio, M. (2006). Rap1: a turnabout for the crosstalk betweencadherins and integrins. Eur. J. Cell Biol. 85, 283-293.

Rozario, T. and DeSimone, D. W. (2010). The extracellular matrix in development andmorphogenesis: a dynamic view. Dev. Biol. 341, 126-140.

Rozario, T., Dzamba, B., Weber, G. F., Davidson, L. A. and DeSimone, D. W. (2009).The physical state of fibronectin matrix differentially regulates morphogeneticmovements in vivo. Dev. Biol. 327, 386-398.

Sahai, E. and Marshall, C. J. (2002). ROCK and Dia have opposing effects on adherensjunctions downstream of Rho. Nat. Cell Biol. 4, 408-415.

Sakai, T., Larsen, M. and Yamada, K. M. (2003). Fibronectin requirement in branchingmorphogenesis. Nature 423, 876-881.

Sallee, J. L., Wittchen, E. S. and Burridge, K. (2006). Regulation of cell adhesion byprotein-tyrosine phosphatases: II. Cell-cell adhesion. J. Biol. Chem. 281, 16189-16192.

Sangrar, W., Gao, Y., Scott, M., Truesdell, P. and Greer, P. A. (2007). Fer-mediatedcortactin phosphorylation is associated with efficient fibroblast migration and isdependent on reactive oxygen species generation during integrin-mediated cell adhesion.Mol. Cell. Biol. 27, 6140-6152.

Schwartz, E. A., Bizios, R., Medow, M. S. and Gerritsen, M. E. (1999). Exposure ofhuman vascular endothelial cells to sustained hydrostatic pressure stimulatesproliferation. Involvement of the alphaV integrins. Circ. Res. 84, 315-322.

Schwartz, M. A. and Assoian, R. K. (2001). Integrins and cell proliferation: regulationof cyclin-dependent kinases via cytoplasmic signaling pathways. J. Cell Sci. 114, 2553-2560.

Shay-Salit, A., Shushy, M., Wolfovitz, E., Yahav, H., Breviario, F., Dejana, E. andResnick, N. (2002). VEGF receptor 2 and the adherens junction as a mechanicaltransducer in vascular endothelial cells. Proc. Natl. Acad. Sci. USA 99, 9462-9467.

Shewan, A. M., Maddugoda, M., Kraemer, A., Stehbens, S. J., Verma, S., Kovacs, E.M. and Yap, A. S. (2005). Myosin 2 is a key Rho kinase target necessary for the localconcentration of E-cadherin at cell-cell contacts. Mol. Biol. Cell 16, 4531-4542.

Sonnenberg, A. and Liem, R. K. (2007). Plakins in development and disease. Exp. CellRes. 313, 2189-2203.

Stoker, A. W. (2005). Protein tyrosine phosphatases and signalling. J. Endocrinol. 185,19-33.

Symowicz, J., Adley, B. P., Gleason, K. J., Johnson, J. J., Ghosh, S., Fishman, D. A.,Hudson, L. G. and Stack, M. S. (2007). Engagement of collagen-binding integrinspromotes matrix metalloproteinase-9-dependent E-cadherin ectodomain shedding inovarian carcinoma cells. Cancer Res. 67, 2030-2039.

Theveneau, E., Marchant, L., Kuriyama, S., Gull, M., Moepps, B., Parsons, M. andMayor, R. (2010). Collective chemotaxis requires contact-dependent cell polarity. Dev.Cell 19, 39-53.

Todorovic, V., Desai, B. V., Schroeder Patterson, M. J., Amargo, E. V., Dubash, A.D., Yin, T., Jones, J. C. and Green, K. J. (2010). Plakoglobin regulates cellmotility through Rho- and fibronectin-dependent Src signaling. J. Cell Sci. 123,3576-3586.

Trepat, X., Wasserman, M. R., Angelini, T. E., Millet, E., Weitz, D. A., Butler, J. P.and Fredberg, J. J. (2009). Physical forces during collective cell migration. Nat. Phys.5, 426-430.

Tsuruta, D. and Jones, J. C. (2003). The vimentin cytoskeleton regulates focal contactsize and adhesion of endothelial cells subjected to shear stress. J. Cell Sci. 116, 4977-4984.

Tzima, E., Irani-Tehrani, M., Kiosses, W. B., Dejana, E., Schultz, D. A., Engelhardt,B., Cao, G., DeLisser, H. and Schwartz, M. A. (2005). A mechanosensory complexthat mediates the endothelial cell response to fluid shear stress. Nature 437, 426-431.

Van Aelst, L. and Symons, M. (2002). Role of Rho family GTPases in epithelialmorphogenesis. Genes Dev. 16, 1032-1054.

Walker, J. L., Menko, A. S., Khalil, S., Rebustini, I., Hoffman, M. P., Kreidberg, J.A. and Kukuruzinska, M. A. (2008). Diverse roles of E-cadherin in the morphogenesisof the submandibular gland: insights into the formation of acinar and ductal structures.Dev. Dyn. 237, 3128-3141.

Wang, H., Radjendirane, V., Wary, K. K. and Chakrabarty, S. (2004). Transforminggrowth factor beta regulates cell-cell adhesion through extracellular matrix remodelingand activation of focal adhesion kinase in human colon carcinoma Moser cells. Oncogene23, 5558-5561.

Wang, X., He, L., Wu, Y. I., Hahn, K. M. and Montell, D. J. (2010). Light-mediatedactivation reveals a key role for Rac in collective guidance of cell movement in vivo.Nat. Cell Biol. 12, 591-597.

Warner, S. J. and Longmore, G. D. (2009). Cdc42 antagonizes Rho1 activity at adherensjunctions to limit epithelial cell apical tension. J. Cell Biol. 187, 119-133.

Watanabe, T., Sato, K. and Kaibuchi, K. (2009). Cadherin-mediated intercellularadhesion and signaling cascades involving small GTPases. Cold Spring Harb. Perspect.Biol. 1, a003020.

Watt, F. M. (2002). Role of integrins in regulating epidermal adhesion, growth anddifferentiation. EMBO J. 21, 3919-3926.

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 MLCKregulates adhesion disassembly. Nat. Cell Biol. 6, 154-161.

Wildenberg, G. A., Dohn, M. R., Carnahan, R. H., Davis, M. A., Lobdell, N. A.,Settleman, J. and Reynolds, A. B. (2006). p120-catenin and p190RhoGAP regulatecell-cell adhesion by coordinating antagonism between Rac and Rho. Cell 127, 1027-1039.

Wilson, E., Sudhir, K. and Ives, H. E. (1995). Mechanical strain of rat vascular smoothmuscle cells is sensed by specific extracellular matrix/integrin interactions. J. Clin.Invest. 96, 2364-2372.

Yamada, S. and Nelson, W. J. (2007). Localized zones of Rho and Rac activities driveinitiation and expansion of epithelial cell-cell adhesion. J. Cell Biol. 178, 517-527.

Yano, H., Mazaki, Y., Kurokawa, K., Hanks, S. K., Matsuda, M. and Sabe, H. (2004).Roles played by a subset of integrin signaling molecules in cadherin-based cell-celladhesion. J. Cell Biol. 166, 283-295.

Yin, T., Getsios, S., Caldelari, R., Kowalczyk, A. P., Muller, E. J., Jones, J. C. andGreen, K. J. (2005). Plakoglobin suppresses keratinocyte motility through both cell-cell adhesion-dependent and -independent mechanisms. Proc. Natl. Acad. Sci. USA 102,5420-5425.

Yonemura, S., Wada, Y., Watanabe, T., Nagafuchi, A. and Shibata, M. (2010). alpha-Catenin as a tension transducer that induces adherens junction development. Nat. CellBiol. 12, 533-542.

Zhong, C., Kinch, M. S. and Burridge, K. (1997). Rho-stimulated contractility contributesto the fibroblastic phenotype of Ras-transformed epithelial cells. Mol. Biol. Cell 8,2329-2344.

Zhurinsky, J., Shtutman, M. and Ben-Ze’ev, A. (2000). Plakoglobin and beta-catenin:protein interactions, regulation and biological roles. J. Cell Sci. 113, 3127-3139.

1193Adhesive crosstalk

Jour

nal o

f Cel

l Sci

ence