Top Banner
Genetic Analyses of Integrin Signaling Sara A. Wickstro ¨m 1 , Korana Radovanac 2 , and Reinhard Fa ¨ ssler 2 1 Paul Gerson Una Group, Skin Homeostasis and Ageing, Max Planck Institute for Biology of Ageing, 50937 Cologne, Germany 2 Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany Correspondence: [email protected]; [email protected] The development of multicellular organisms, as well as maintenance of organ architecture and function, requires robust regulation of cell fates. This is in part achieved by conserved signaling pathways through which cells process extracellular information and translate this information into changes in proliferation, differentiation, migration, and cell shape. Gene deletion studies in higher eukaryotes have assigned critical roles for components of the extracellular matrix (ECM) and their cellular receptors in a vast number of developmental processes, indicating that a large proportion of this signaling is regulated by cell-ECM inter- actions. In addition, genetic alterations in components of this signaling axis play causative roles in several human diseases. This review will discuss what genetic analyses in mice and lower organisms have taught us about adhesion signaling in development and disease. A lmost all cells in multicellular organisms are surrounded by a three-dimensional organ- ized meshwork of macromolecules that consti- tute the extracellular matrix (ECM). The ECM is a dynamic structure that is generated and constantly remodeled by cells that secrete and manipulate its components into a precise con- figuration. It functions as a structural frame- work that provides cells with positional and environmental information, but also forms specialized structures such as cartilage, tendons, basement membranes (BM), bone, and teeth. In addition to its structural properties, the ECM acts as a signaling platform that regulates a large number of cellular functions. It is capa- ble of binding growth factors, chemokines, and cytokines thereby modulating their bio- availability and activity. On the other hand, the ECM is recognized by multiple cell surface receptors that transmit information from the extracellular environment by propagating intra- cellular signals (for a review, see Hynes 2009). The major cell surface receptors that recog- nize and assemble the ECM are integrins. Integ- rins are heterodimeric transmembrane proteins composed of a and b subunits. Eighteen a sub- units and eight b subunits can assemble in 24 different combinations with overlapping sub- strate specificity and cell-type-specific expres- sion patterns (Hynes 2002; Humphries et al. 2006). This, together with the ability of different heterodimers to assemble specific intracellular signaling complexes, provides multiple layers of signaling specificity to these receptors. Con- versely, the integrin expression profile of a given cell type determines which ECM components it Editors: Richard Hynes and Kenneth Yamada Additional Perspectives on Extracellular Matrix Biologyavailable at www.cshperspectives.org Copyright # 2011 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a005116 Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116 1 on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/ Downloaded from
23

Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

Feb 26, 2021

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

Genetic Analyses of Integrin Signaling

Sara A. Wickstrom1, Korana Radovanac2, and Reinhard Fassler2

1Paul Gerson Una Group, Skin Homeostasis and Ageing, Max Planck Institute for Biology of Ageing,50937 Cologne, Germany

2Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany

Correspondence: [email protected]; [email protected]

The development of multicellular organisms, as well as maintenance of organ architectureand function, requires robust regulation of cell fates. This is in part achieved by conservedsignaling pathways through which cells process extracellular information and translate thisinformation into changes in proliferation, differentiation, migration, and cell shape. Genedeletion studies in higher eukaryotes have assigned critical roles for components of theextracellular matrix (ECM) and their cellular receptors in a vast number of developmentalprocesses, indicating that a large proportion of this signaling is regulated by cell-ECM inter-actions. In addition, genetic alterations in components of this signaling axis play causativeroles in several human diseases. This review will discuss what genetic analyses in miceand lower organisms have taught us about adhesion signaling in development and disease.

Almost all cells in multicellular organisms aresurrounded by a three-dimensional organ-

ized meshwork of macromolecules that consti-tute the extracellular matrix (ECM). The ECMis a dynamic structure that is generated andconstantly remodeled by cells that secrete andmanipulate its components into a precise con-figuration. It functions as a structural frame-work that provides cells with positional andenvironmental information, but also formsspecialized structures such as cartilage, tendons,basement membranes (BM), bone, and teeth.In addition to its structural properties, theECM acts as a signaling platform that regulatesa large number of cellular functions. It is capa-ble of binding growth factors, chemokines,and cytokines thereby modulating their bio-availability and activity. On the other hand,

the ECM is recognized by multiple cell surfacereceptors that transmit information from theextracellular environment by propagating intra-cellular signals (for a review, see Hynes 2009).

The major cell surface receptors that recog-nize and assemble the ECM are integrins. Integ-rins are heterodimeric transmembrane proteinscomposed of a and b subunits. Eighteen a sub-units and eight b subunits can assemble in 24different combinations with overlapping sub-strate specificity and cell-type-specific expres-sion patterns (Hynes 2002; Humphries et al.2006). This, together with the ability of differentheterodimers to assemble specific intracellularsignaling complexes, provides multiple layersof signaling specificity to these receptors. Con-versely, the integrin expression profile of a givencell type determines which ECM components it

Editors: Richard Hynes and Kenneth Yamada

Additional Perspectives on Extracellular Matrix Biology available at www.cshperspectives.org

Copyright # 2011 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a005116

Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116

1

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 2: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

can bind. Signals arising from integrins regulatevirtually all aspects of cell behavior, includingcell migration, survival, cell cycle progression,and differentiation.

Genetics has proven to be a powerful tool todissect the functions of ECM–cell interactionsin complex organisms. To date, all of the integ-rin subunits and their major ligands have beendeleted in mice. Given the large variety of cellu-lar processes regulated by adhesion signaling,it is not surprising that a significant subset ofthese proteins has proven to be essential forembryonic development and/or tissue mainte-nance. However, in addition to underlining theimportance of cell-ECM interactions in devel-opment, genetic studies also revealed criticalroles for tissue- and cell-type-specific modesof adhesion signaling and provided importantinsights into human disease.

THE INTEGRIN-INTERMEDIATEFILAMENT AXIS

Basement membranes (BMs) are dense sheets ofextracellular matrix that function as structuralbarriers that separate epithelial and endothelialcells as well as peripheral nerve axons, fat cells,and muscle cells from the underlying tissuestroma. BMs provide structural support, separatetissues into compartments, and regulate cellbehavior. All cell types are known to producecomponents of BMs, which include type IV col-lagen, laminin, fibronectin, heparan sulfateproteoglycans, and nidogen/entactin as well asseveral minorcomponents (Erickson andCouch-man 2000; Yurchenco 2010). The molecularcom-position of the BM varies among different tissues,conferring signaling specificity important fordefining the specialized functions of epithelialand endothelial cells in different organs.

All BMs contain laminins, a family of 16heterotrimeric glycoproteins generated by thecombination of 5a, 4b, and 3g chains. Whenpresent in sufficient concentrations, lamininnetworks can self-assemble into polymers thatinteract with other ECM components, as wellas cell surface receptors such as integrins anddystroglycan. The laminin isoforms presentin BMs are regulated in a tissue-specific

manner, as well as temporally during develop-ment (Miner 2008; Yurchenco 2010). The cen-tral role of laminins in BM assembly isillustrated by gene deletion studies in mice.Laminin-111 (containing a1, b1, and g1chains) and laminin-511 (containing a5, b1,and g1 chains) are the central constituents ofperi-implantation stage BMs. Mice deficient ing1 or b1 subunits are unable to generate lami-nin trimers, and therefore lack BMs (Smythet al. 1999; Miner et al. 2004).

Laminin-332 (containing a3, b3, and g2chains; previously termed laminin-5) is a com-ponent of epithelial BMs and thus present inskin, stratified squamous mucosa, amnion, andcornea. Its main task is to maintain epithelialintegrity and epithelial-mesenchymal cohesionin tissues exposed to high mechanical forces.This function is facilitated by the uniqueability of laminin-322 to interact with two dis-tinct integrin heterodimers. Its interaction witha3b1 integrin results in the assembly of canonicalfocal adhesions (FA), whereas the interactionwith a6b4 integrin results in the formation ofspecialized adhesion complexes termed hemides-mosomes (Tsuruta et al. 2008).

Hemidesmosomes—Structure and Assembly

Hemidesmosomes were first characterized at theultrastructural level as electron-dense clusters atthe plasma membrane-ECM interphase (Weissand Ferris 1954). Further studies identifiedthem as multiprotein adhesion complexes pre-sent in stratified and simple epithelia. Two typesof hemidesmosomes can be distinguished basedon their molecular composition. Type I or clas-sical hemidesmosomes are found in stratifiedepithelium such as the skin and contain threetransmembrane proteins: a6b4 integrin, tetra-spanin CD151, and type XVII collagen (alsocalled bullous pemphigoid [BP] antigen 180)(Fig. 1). Type II hemidesmosomes are foundin simple epithelia such as intestine, and containonly integrin a6b4 (Uematsu et al. 1994;reviewed in Litjens et al. 2006).

The unique feature of hemidesmosomesis that they connect the ECM to the intermedi-ate filament (IF) network. This interaction is

S.A. Wickstrom et al.

2 Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 3: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

established by two plakin proteins, plectins andBP230 (also called BPAG1), of which plectin ispresent both in type I and II hemidesmosomes.Plectins are large cytoplasmic proteins, whichat their carboxyl terminus contain six plakinrepeats. A stretch of basic residues linking thefourth and fifth plakin repeat mediates theinteraction of plectin with IFs. The amino ter-minus contains two calponin-homology (CH)domains that constitute an actin-binding do-main. Through the actin-binding domain,plectins can associate with either the cytoplas-mic domain of the b4 integrin subunit or actinfilaments in a mutually exclusive manner, which

explains the absence of actin in hemides-mosomes (Rezniczek et al. 1998; Geerts et al.1999; Litjens et al. 2003). The interaction ofplectin with the cytoplasmic tail of b4 integrinis considered as the initial step of hemidesmo-some assembly (Geerts et al. 1999; Koster et al.2004). The cytoplasmic tail of b4 integrin isunusually large and shares no homology withother b integrin subunits. Its interaction withplectin has been shown to induce a conforma-tional change in the integrin tail (de Peredaet al. 2009). Because the recruitment of typeXVII collagen and the plakin BP230 to hemides-mosomes requires prior interaction of plectin

Stable adhesion

Cell migrationsignaling

Hemidesmosomedisassembly

Collagen XVII

Plectin

BP230

IF

Basalkeratinocyte

CD151

LM 322

α6 Integrinβ4 Integrin

Lamina lucida

Lamina densa

BM

P - S1364P - S1360

P - S1356

Figure 1. Molecular architecture of type I hemidesmosomes. Schematic depiction of a type I hemidesmosomefound in stratified epithelia such as in basal skin keratinocytes. The core component is a6b4 integrin, whichbinds the basement membrane (BM) component laminin (LM)-322. a6b4 integrin recruits the plakin proteinplectin through multiple interactions with the b4 integrin cytoplasmic tail, which initiates the formation ofhemidesmosomes. This is followed by the recruitment of collagen XVII, which interacts both with b4 integrinand plectin as well as with LM 322. Collagen XVII in turn mediates the recruitment of another plakin, bullouspemphigoid antigen 230 (BP 230), which together with plectin provides the connection to intermediate fila-ments (IF). This linkage is essential to stabilize the hemidesmosome and to provide stable adhesion of the basalkeratinocyte to the BM. Also, the transmembrane protein tetraspanin CD151 that interacts with a6 integrin isfound in type I hemidesmosomes. Phosphorylation of serines (S) 1356, 1360, and 1364 on the cytoplasmic tail ofb4 integrin by growth factors induces disassembly of hemidesmosomes, which promotes cell migration and sig-naling. The molecules are not drawn to scale.

Genetic Analyses of Integrin Signaling

Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116 3

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 4: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

with b4, this conformational change mightfacilitate the interactions of both proteins withthe integrin (Koster et al. 2004).

Deletion of a6b4 leads to the loss of hemi-desmosomes and epithelial detachment in mice(Dowling et al. 1996; van der Neut et al. 1996),indicating that BP180, despite its ability to bindlaminin-332 and plectin in vitro, is not suffi-cient to maintain adhesion of cells to laminin-332 in the absence of a6b4 (Tasanen et al.2004) and identifying a6b4 as the core compo-nent of hemidesmosomes. Another criticalevent in hemidesmosome assembly is the inter-action between b4 integrin tails and plectin.Blocking this interaction compromises hemi-desmosome assembly in vitro (Geerts et al.1999; Koster et al. 2001). In plectin-deficientmice hemidesmosomes appear ultrastructurallynormal, but their number and mechanical sta-bility are reduced (Andra et al. 1997). The factthat type II hemidesmosomes lack both typeXVII collagen and BP230 further shows thata6b4 and plectin alone are sufficient to initiatethe formation of these structures and to main-tain their integrity.

Although a6b4 is the only integrin foundin hemidesmosomes, other integrins can indi-rectly contribute to their assembly. Integrina3b1 has been shown to cluster in “pre-hemi-desmosomal” structures on the basal cell surfaceof human keratinocytes together with CD151.However, as hemidesmosomes mature, a3b1integrins become recruited to cell-cell or focalcontacts, whereas CD151 remains in the hemi-desmosomes (Sterk et al. 2000). The initiala3b1-containing adhesions have been pro-posed to contribute to the recruitment of a6b4to the plasma membrane, and thereby increasethe efficacy with which hemidesmosomes areformed. This is supported by the observationthat b1-deficient mice have reduced numbersof hemidesmosomes in the skin, although itshould be noted that the expression level ofa6b4 is also reduced in these mice, suggestingthat the mechanism could also be more indirect(Brakebusch et al. 2000). Polarity proteinshave also been shown to play a role in hemides-mosome assembly through the regulation ofpolarized targeting of integrins. In the larval

epidermis of the zebrafish, the basolateralidentity protein Lgl2 is required for hemides-mosome formation through recruitment of b4integrin to the basal plasma membrane (Sona-wane et al. 2005, 2009). Taken together, theformation of hemidesmosomes requires theestablishment of a basolateral membrane iden-tity through polarity proteins and possibly alsoother integrin-based adhesion structures to re-cruit a6b4 integrin to these sites. a6b4 integrinthen interacts with BM laminin and togetherwith plectin nucleates stable, IF-bound hemi-desmosomes. Interestingly, however, keratino-cytes expressing a b4 integrin subunit unableto bind laminin can still form structures thatcontain all components of type I hemides-mosomes, which implies that ligand bindingis not necessary for their formation (Nieverset al. 1998, 2000). These complexes, however, dif-fer in their morphology, density, and dynamicsfrom normal hemidesmosomes, suggesting thatthe integrin-laminin interaction plays a role inthe stability and structural organization of hemi-desmosomes (Geuijen and Sonnenberg 2002).

Hemidesmosomes—Guardians ofEpidermal Integrity

A central function of the skin is to act as a bidi-rectional barrier to prevent dehydration and toprotect against injury and pathogens. In addi-tion, it is exposed to constant mechanical stress.On the other hand, this tissue is continuouslyrenewed through a terminal differentiation pro-gram, in which basal keratinocytes detach fromthe BM and move to the upper layers of the epi-thelium where they slough off. Finally, in caseof injury, keratinocytes need to rapidly acquirea motile phenotype and migrate to close thewound. These functions require mechanicallystable but dynamic modes of cell adhesion.Hemidesmosomes are found in the basal plasmamembrane of basal keratinocytes, where theyfunction to attach these cells firmly to the BMseparating the epidermis from the dermis (Fig.1). Interestingly, a6b4 integrins and thus alsohemidesmosomes are not required for skinmorphogenesis or developmental homeostasis(DiPersio et al. 2000). The importance of

S.A. Wickstrom et al.

4 Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 5: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

hemidesmosomes in maintaining epidermalintegrity is, however, shown by multiple linesof genetic evidence. Ablation of genes encodinga6 integrin,b4 integrin, or plectin in mice resultsin severe blistering of the skin causing neonataldeath because of a severe epithelial barrier defect(Dowling et al. 1996; Georges-Labouesse et al.1996a; van der Neut et al. 1996; Andra et al.1997). In line with their dispensable role in hemi-desmosome assembly, mice lacking type XVIIcollagen or BP230 display only mild forms ofskin blistering (Guo et al. 1995; Nishie et al.2007), whereas deletion of CD151 is dispensablefor both hemidesmosome stability and the integ-rity of skin (Wright et al. 2004).

No experimental data exist on the dynamicbehavior of hemidesmosomes in vivo, but it hasbeen analyzed in keratinocyte culture, in whichhemidesmosomes have been shown to displaya certain dynamics also in nonmigratory cells(Tsuruta et al. 2003). When cells are stimu-lated to migrate, a6b4 integrin is mobilizedfrom hemidesmosomes to actin-based struc-tures such as lamellipodia and filopodia to pro-mote cell motility, resulting in hemidesmosomedisassembly (Geuijen and Sonnenberg 2002;Tsuruta et al. 2003). This type of relocalizationhas also been visualized in human wound edges,where b4 integrins can be seen in trailing-edgehemidesmosomes as well as in lamellipodia ofthe leading edge (Underwood et al. 2009). Invitro analyses have shown that the mobilizationof a6b4 integrin from hemidesmosomes isfacilitated by growth factor-mediated phos-phorylation of multiple residues in the cyto-plasmic tail of b4. Both serines and tyrosinescan be phosphorylated, but the exact sites andtheir relevance remain controversial. The cur-rent data suggest that serine phosphorylationmight be more relevant under physiologicalconditions, leading to hemidesmosome disas-sembly through the release of plectin from theintegrin (Margadant et al. 2008) (Fig. 1)

a6b4 Integrin and Cancer

b4 integrin was originally identified as a “tu-mor-specific” protein (Tumor Surface Protein180) up-regulated in metastatic variants of

mouse lung carcinoma and melanoma cell lines(Kennel et al. 1981), and has later been shown tobe up-regulated in several human cancers, sug-gesting that the expression ofb4 integrin is ben-eficial for tumor cells (Giancotti 2007). The roleof a6b4 integrin signaling in tumorigenesishas been studied in mice carrying a truncatedcytoplasmic tail of b4 lacking the tyrosinephosphorylation sites as well as other potentialinteraction motifs. In contrast to mice lackingthe entire b4 cytoplasmic domain, whichdisplay extensive skin blistering because of theabsence of hemidesmosomes (Murgia et al.1998), mice with a more restricted truncationdo not show these defects. However, they showdelayed wound healing and defective neoangio-genesis in tumor xenografts (Nikolopoulos et al.2004, 2005). In addition, when crossed to theMMTV-Neu mice that carry an activated formof epidermal growth factor receptor-2 (ErbB2)driven by a mouse mammary tumor virus(MMTV) promoter leading to mammary tu-mors, the b4 mutant mice display delayedtumor onset, impaired tumor growth and de-creased metastatic potential (Guo et al. 2006).Whether this applies to other tumor types aswell remains to be shown.

The tumor-promoting properties of a6b4integrin seem to result from both promigratoryand signaling functions. b4 integrin has beenshown to cross-talk with several receptor tyro-sine kinases (RTK), including ErbB2, epidermalgrowth factor receptor (EGF-R), Met and Ron(Mariotti et al. 2001; Trusolino et al. 2001;Santoro et al. 2003; Guo et al. 2006). Followingstimulation, these RTKs induce activation ofphosphatidylinositol 3-kinase (PI3-K) or Srcfamily kinases (SFK), leading to phosphoryla-tion of theb4 integrin tail, disassembly of hemi-desmosomes, and induction of cell motility(Mariotti et al. 2001; Santoro et al. 2003). Onthe other hand, b4 integrin has been shown topromote SFK-dependent phosphorylation ofthe catalytic sites of RTKs and their substrates(Bertotti et al. 2006; Guo et al. 2006), therebyamplifying their signaling ability. Takentogether, a6b4 integrin seems to have a proin-vasive and proangiogenic activity in tumors,but additional genetic studies are needed to

Genetic Analyses of Integrin Signaling

Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116 5

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 6: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

determine whether this role is limited to malig-nancies involving hyperactivation of specificRTKs. In addition, several groups have failedto detect significant tyrosine phosphorylationof b4 integrin in keratinocytes or transformedcell lines in response to stimulation with EGFand hemidesmosome disassembly, making therelevance of tyrosine phosphorylation eventscontroversial (Rabinovitz et al. 1999, 2004; Altet al. 2001; Wilhelmsen et al. 2007). Knockinmice carrying mutations in specific phosphory-lation sites of the b4 integrin subunit wouldprovide more conclusive information on thein vivo significance of the various phosphoryla-tion events in a6b4 integrin signaling, hemi-desmosome turnover, and invasion.

THE INTEGRIN-ACTIN AXIS

In contrast to a6b4, most integrins engage theactin cytoskeleton following ligand binding. Acentral ligand of actin-associated integrins isFN, which is recognized by 11 integrin hetero-dimers in mice and humans, and it will be usedin this section as an example to discuss the prin-ciples of ECM-integrin interactions leading toengagement and remodeling of the actin cytoske-leton. FN is a large, modular glycoprotein thatexists in two forms; cellular FN which is presentin tissues where it is assembled into a fibrillarmatrix, and plasma FN, which is produced byhepatocytes and is secreted into the blood whereit remains in a nonfibrillar, soluble form, or fol-lowing entry into tissues becomes incorporatedinto the ECM (Leiss et al. 2008). FN is foundonly invertebrates, and it has co-evolved togetherwith the cardiovascular system. Consistently, FNis critical for the development of the vasculature,where it localizes between the endothelium andperivascular cells. Deletion of FN in mice resultsin embryonic lethal cardiovascular defects, whichvary depending on the genetic background.FN-null embryos from 129S4 mice are unableto form the dorsal aorta, indicative of an earlydefect already in vasculogenesis, whereas thisstructure is present in C57BL/6 derived embryosthat display defects in vascular lumen formation(George et al. 1993, 1997; Georges-Labouesseet al. 1996b).

FN is secreted as a disulfide-bonded dimer,and its deposition into a fibrillar matrix is acell-driven process that critically depends onintegrins. So fara5b1, avb3, a4b1, and aIIbb3integrins have been shown to induce fibri-llogenesis in vitro. These integrins bind FNthat is secreted as a compact globular structurewhere binding sites for other FN molecules areburied within the protein (see Schwarzbauerand DeSimone 2011). Binding is followed bythe activation of integrin signaling, leading tothe recruitment of cytoplasmic proteins toform focal complexes (FCs). Several FC com-ponents are actin-binding and modulatoryproteins, allowing the recruitment and reorgan-ization of the actin cytoskeleton at these sitesand their maturation into FAs (Geiger andYamada 2011). The major FN-fibril-formingintegrin a5b1 leaves FAs and moves along F-actin to the cell center to form fibrillar adhe-sions and to facilitate the generation of mechan-ical tension via the actin cytoskeleton, leading tostretching of the bound FN molecule, unravel-ing of the cryptic self-association sites, andfinally the binding to other FN moleculesresulting in fibril formation (Mao and Schwarz-bauer 2005; Leiss et al. 2008; Schwarzbauer andDeSimone 2011).

Interactions via the RGD Motif

Cell adhesion to FN critically depends on theArg-Gly-Asp (RGD) motif of FN, which is rec-ognized by a5b1, the av family, a8b1, a9b1,and the platelet-specific aIIbb3 integrins(Fig. 2). a5b1 is considered the major integrinresponsible for FN assembly, and its interactionwith the RGD motif is required for this func-tion. However, although deletion of a5 integrinin mice also leads to embryonic lethality andvascular defects, these mice develop signifi-cantly further than the FN-null mice (Yanget al. 1993). This is attributable to the abilityof cells to assemble FN using other integrins,mainly of the av subfamily. The deletion of allfive av heterodimers also leads to late embry-onic defects in vascular development mainlyin the placenta. A subset of animals can evenproceed through embryonic development and

S.A. Wickstrom et al.

6 Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 7: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

die after birth because of hemorrhages, indicat-ing that, unlike a5, av integrins are dispensablefor vasculogenesis and partly also for angiogen-esis (Bader et al. 1998). Only a double knockoutof the av and a5 integrin genes results in a lossof FN fibrillogenesis (Yang et al. 1999). Interest-ingly, although the RGD motif is central for theinteraction of FN with a5b1 and avb3, theinactivation of this motif by a RGD to RGEpoint mutation also allows FN fibrillogenesisin vivo. The knockin mice carrying this muta-tion display a phenotype closely resemblingthat of the a5-null mice (Takahashi et al.2007). This highlights the importance of thismotif in FN signaling through a5b1, but alsoshows the ability of other integrin interactionsites on FN to take over the role of RGD in FNfibrillogenesis. Whether these sites play a rolein FN assembly when the RGD sequence isintact, remains open. Together, these resultsindicate that FN fibrillogenesis is not sufficientfor it to carry out its functions during develop-ment. They further illustrate that the majority of

FN signaling in embryogenesis occurs througha5b1 and that av and a5b1 integrins relaydistinct signals on FN binding. The severity ofthe FN-null phenotype in contrast to the integ-rin deletions also suggests that the functionsof FN during development extend beyondthe FN-integrin signaling axis. These functionscould be related to its mechanical role in theECM, or its ability to induce integrin-indepen-dent signaling, for example, through sequester-ing growth factors such as vascular endothelialgrowth factor (VEGF) or transforming growthfactor-b (TGF-b).

Non-RGD Binding Integrins

The FN gene can be alternatively spliced allow-ing the expression of up to 20 possible mo-nomeric isoforms in humans and up to 12 inmouse, potentially giving rise to a larger varietyof FN dimers (Pankov and Yamada 2002). Someof these splice variants can generate additionalintegrin interaction sites on FN. One of them

NGRMotif

Domain

Integrin

Fn1

Fn2

Fn3

Extra domain

Variable region

Reporteddevelopmentalfunction

? Lungmorphogenesis

Lymphangio-genesis

Placentalangiogenesis

SomitogenesisVasculogenesisAngiogenesis

Plateletaggregation

Lymphaticvalvemorphogenesis(α9β1)

Vascular remodellingPericyte recruitment

α4β1α4β7

α4β1

NH2

RGD EDGIHELLDVREDV

S S

COOH

α9β1αIIβ3α9β1α8β1 α5β1

αvβ5αvβ6

α5β1αvβ3

αvβ1αvβ3

Figure 2. Developmental functions of fibronectin-integrin interactions. Fibronectin (FN) is a dimeric glycyo-protein consisting of type I, type II, and type III modular repeats. Dimerization is achieved by disulfide bondsmediated by the two cysteines (S) in the COOH-terminus of the protein. The binding domains and motifs forintegrins, as well as mouse developmental functions reported to depend on the particular interaction are indi-cated. Integrins that have been shown to mediate FN fibrillogenesis in vitro are marked by gray boxes.

Genetic Analyses of Integrin Signaling

Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116 7

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 8: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

is the variable region (v-region) that can inter-act with two non-RGD-binding integrins, a4b1and a4b7 (Wayner et al. 1989; Guan and Hynes1990; Fig. 2). These integrins are mainly ex-pressed by hematopoietic cells, although a4b1is also found in several other cell types, suchas neural crest cells and cellular componentsof the cardiovascular and peripheral nervoussystems. The two a4 integrins have many im-portant roles during development. Deletion ofthe a4 integrin gene results in embryonic le-thality because of early placental and cardiovas-cular defects (Yang et al. 1995). However, thesedefects are probably not attributed to the FN-a4b1/a4b7 integrin interaction, as these twointegrins also bind cell counter receptors suchas vascular-cell adhesion molecule-1 (VCAM-1)and MadCAM, respectively. The developmen-tal defects in the a4 integrin-null mice arevery similar to those found in mice lackingVCAM-1, indicating that the observed defectsare caused by an abrogated interaction betweena4b1 integrin and VCAM-1 (Kwee et al. 1995;Yang et al. 1995). From a clinical point of view,it is particularly interesting that the interactionbetween a4b1 and VCAM-1 is important forthe homing of autoreactive T cells to the centralnervous system during the pathogenesis of anautoimmune disease called multiple sclerosis(Yednock et al. 1992; Vajkoczy et al. 2001; Baueret al. 2009). This disease is characterized byaxon demyelination and damage, leading tonumbness, weakness, paresis as well as cogni-tive problems. Blocking antibodies against a4integrin have proven to be an effective treatmentof multiple sclerosis (Steinman 2009), andgenetic analysis in mice showed that a4b1 isrequired for the arrest of T cells on the brainendothelial surface (Bauer et al. 2009).

a4b1 integrins can also interact with thealternative spliced extra domain A (EDA) of FN(Fig. 2), which is highly expressed during de-velopment and during pathological conditionssuch as tumorigenesis. This “reactivation” ofthe embryonic splice pattern is interesting, asa4b1 has recently been shown to play a criti-cal role in tumor lymphangiogenesis in a FN-dependent manner. Both a4b1 and FN arehighly expressed in lymphatic endothelial cells

of tumors, whereas other integrin subunitsseem not to be up-regulated (Garmy-Susiniet al. 2010). Whether this function reallydepends on the EDA domain was, however,not analyzed. In addition, it is not clear whethera4b1 integrin is also regulating developmentallymphangiogenesis, as the constitutive knock-out mice die before the initiation of this process.The EDA domain itself, however, plays a rolein developmental lymphangiogenesis throughintegrin a9b1. Interestingly, this integrin ishighly expressed in lymphatic but not bloodendothelial cells (Huang et al. 2000). Deletionof a9 integrin gene leads to a severe chylothoraxcaused by defects in lymphatic valve morpho-genesis and subsequent respiratory failure(Huang et al. 2000; Bazigou et al. 2009). Thisphenotype was partially recapitulated by dele-tion of EDA. Furthermore, both a9b1 integrinas well as the EDA domain were shown to becritical for FN assembly in lymphatic endothe-lial cells in vitro (Bazigou et al. 2009), in con-trast with previous studies showing that EDAis not required for FN assembly or mouse devel-opment because of compensation by EDB (Tanet al. 2004; Astrof et al. 2007). This study sug-gests that matrix assembly might be regulatedby integrins that are expressed in a tissue-spe-cific manner to allow more stringent controlof distinct anatomical structures.

Linkage to the Actin Cytoskeleton

In addition to catalyzing matrix assembly, thebinding of integrins to FN or other ligands leadsto the induction of multiple modes of intracel-lular signaling. These include activation of clas-sical signaling pathways through tyrosine andserine phosphorylation of specific substrates,resulting ultimately in the regulation of cell sur-vival, growth and differentiation. This signal-ing operates in close collaboration with growthfactor signaling, making it difficult to dissectwhich signals specifically emanate from ligatedintegrins (Legate et al. 2009; Ivaska and Heino2010). The other central signaling mode is therecruitment of filamentous (F-) actin to integ-rin adhesion sites, a process tightly coupled tothe active remodeling of the actin cytoskeleton.

S.A. Wickstrom et al.

8 Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 9: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

The integrin-actin linkage is central to integrinfunction in several respects. First, active actinpolymerization, crosslinking, and subsequentforce generation are critical for the maturationof nascent and short-lived integrin adhesionsinto signaling-competent and stable FAs. Sec-ond, it is required for the precise spatiotemporalcontrol of cell protrusion and retraction duringcell migration. Third, it allows cells to adopt orchange their characteristic shape, for exampleto polarize. Finally, it is critical for FN matrixassembly. Integrins do not bind actin directly,but regulate this linkage by recruiting a largenumber of actin-binding and regulatory pro-teins. A recent study based on database miningidentified 156 signaling, structural and adaptormolecules that can be found in integrin adhe-sions (Zaidel-Bar et al. 2007). Only a small sub-set of these proteins binds directly to integrins,and the large majority is recruited throughprotein-protein interactions between the vari-ous scaffold proteins (for reviews, see Legateand Fassler 2009; Geiger and Yamada 2011). Alarge body of structural, biochemical, andgenetic evidence points to talin, kindlin, andthe integrin-linked kinase (ILK)-pinch-parvin(IPP) complex as some of the central FA com-ponents regulating the integrin-actin linkage.All of these proteins are ubiquitously expressedand regulate a wide range of integrin hetero-dimers. Deletion of any of these proteins leadsto early embryonic lethality, which resultsfrom compromised integrin function anddefects in their connection to the cytoskeleton.On the cell-biological level these defects spanthe entire range of functions attributed to theintegrin-actin linkage. Hence, they can beviewed as global regulators of integrin function.The precise functions of these proteins will bediscussed in the next section.

IN VIVO MECHANISMS OF INTEGRINSIGNALING THROUGH INTRACELLULAREFFECTORS AND SCAFFOLDS

Talin and kindlin are two FA proteins thatdirectly bind integrins and thereby are involvedin the very early events of integrin signaling.They are distinguished from the large group

of FA proteins by their ability to regulate bothintegrin activation (inside-out signaling) andintracellular signaling downstream of ligandbinding (outside-in signaling). Talin is presentin all multicellular eukaryotes; lower eukary-otes encode only a single talin isoform, whereasvertebrates have two talin isoforms, talin-1and talin-2. Talin consists of a large carboxy-terminal rod and an amino-terminal headdomain composed of a FERM domain withfour subdomains: F0, F1, F2, and F3. The F3subdomain contains a phosphotyrosine-bind-ing motif, which harbors a high affinity bindingsite for b-integrin tails. The rod domain con-tains multiple binding sites for the actin–bind-ing protein vinculin as well as for actin itself. Inaddition it contains a second integrin-bindingsite (Critchley and Gingras 2008). The talinhead has been shown to be sufficient for integ-rin binding and activation, whereas the rod isrequired for the scaffolding function of talinin outside-in signaling (Garcia-Alvarez et al.2003; Tanentzapf and Brown 2006; Tanentzapfet al. 2006).

Kindlins are an evolutionarily conservedfamily of multidomain proteins, which in mam-mals consists of three members: kindlin-1 (alsoknown as FERMT1), kindlin-2 (also knownas FERMT2 or MIG-2), and kindlin-3 (alsoknown as FERMT3 or URP2). Although en-coded by separate genes, they show identicaldomain architecture and high sequence simi-larity (Ussar et al. 2006). Like talin, kindlinscontain a FERM domain through which theyinteract with b1, b2, and b3 integrins, butthis FERM domain is unique in that it is inter-rupted by a pleckstrin homology (PH) domain,which provides a putative binding motif formembrane lipids.

Consequences of Loss of IntegrinInside-Out Signaling

Integrins are present on the plasma membranein low-, intermediate-, and high-affinity states.Structural studies suggest that a low-affinity stateis characterized by a bent, “closed” conformationof the extracellular domains and a high-affinitystate by an extended, “open” conformation

Genetic Analyses of Integrin Signaling

Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116 9

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 10: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

(Campbell and Humphries 2011). The extendedconformation is thought to be achieved byseparation of the a and b cytoplasmic tailsthrough binding of talin to the b subunit(Moser et al. 2009b; Shattil et al. 2010). Studiesin cultured cells have shown that the bindingof talin-1 to the cytoplasmic domain of theb-integrin subunit is a common step in b1and b3 integrin activation in vitro (Tadokoroet al. 2003). In contrast, kindlin alone is notsufficient to activate these integrins, but itsco-operation with talin is required to generatefully active integrins (Montanez et al. 2008;

Moser et al. 2008, 2009a). The molecular detailsof this cooperation are not clear.

Regulation of the ligand binding affinity isfundamental for various cellular functions.For example, the platelet integrin aIIbb3 needsto be maintained inactive to prevent it frombinding fibrinogen present in the blood streamthus prohibiting platelet aggregation andthrombus formation. On the other hand, thisintegrin needs to be rapidly activated to mediateplatelet adhesion on vascular injury to preventbleeding (Fig. 3). Similarly, leukocytes requireintegrin activation to adhere to and migrate

GPI

1

2

3 4

5

54321

Talin

Kindlin

Adaptor proteins

F-Actin

Resting platelet Inside-out signaling Outside-in signaling Platelet spreading Platelet aggregation

Inactive integrins Talin and kindlinrecruitment

Activation of integrin

Ligation of active integrinsRecruitment of adaptors

Cytoskeletal rearrangementAdhesion strengthening

Binding to soluble fibrinogenThrombus formation

αβ Integrin

Figure 3. Talin and kindlin regulate bidirectional integrin signaling. Bidirectional integrin signaling is essentialfor platelets (tan) to seal the injured blood vessel endothelium (red) and stop bleeding. Integrins in circulating,resting platelets exist in a low affinity state indicated by a bent confirmation (1). Following vessel injury, von Wil-lebrand factor (vWF) and collagen are exposed to bind their receptors GPIb and GPVI that are expressed on thesurface of platelets. Together with locally produced thrombin these receptors trigger the activation of aIIbb3integrin. This is achieved by promoting the association of talin-1 and kindlin-3 with the cytoplasmic tail ofb3 integrin, facilitating a conformational change in the integrin (inside-out signaling) (2). The conformationalchange allows integrins to bind fibrinogen, vWF, and fibronectin with high affinity. As a result, platelets adhere tothe vessel wall. Integrin ligation subsequently initiates signaling through kindlin and talin (outside-in signaling)(3) resulting in the recruitment of adaptor proteins and rearrangement of the cytoskeleton to promote cellspreading (4). This, together with integrin-mediated binding of soluble fibrinogen, results in platelet aggrega-tion and formation of a stable clot (5). The molecules are not drawn to scale.

S.A. Wickstrom et al.

10 Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 11: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

across the endothelium to combat pathogens.The importance of this process is illustrated bythe phenotypes of talin- and kindlin-deficientmice. Deletion of talin-1 or kindlin-3 in plate-lets blocks activation of b1 and b3 integrins,leading to the inability of platelets to bind fibri-nogen and to form clots. As a result, these micesuffer from severe bleeding (Nieswandt et al.2007; Petrich et al. 2007; Moser et al. 2008).Similarly, neutrophils lacking kindlin-3 areunable to activate b2 integrins, resulting inloss of neutrophil adhesion to activated endo-thelial cells (Moser et al. 2009a). Talin has alsobeen shown to bind and activate neutrophilintegrin b2 in vitro (Simonson et al. 2006).The importance of integrin affinity regulationin adherent cell types is not as clear. Althoughdeletion of talin and kindlin in mesenchymalor epithelial cells leads to defects in integrinactivation, these cells are still capable of adher-ing, albeit to a reduced extent (Ussar et al.2008; Zhang et al. 2008), suggesting that underconditions of less stringent requirements forrapid induction of adhesion, high concentra-tions of ligands, and the absence of high shearflow, avidity regulation might be the predomi-nant mechanism of integrin regulation. Thefact that both talin and kindlin are also impor-tant in regulation of outside-in signaling makesthe in vivo dissection of these two processesdifficult.

Consequences of Loss of Outside-InSignaling

A central piece of evidence that talin has impor-tant functions besides integrin activation comesfrom studies performed with talin-null flies,which show that in the absence of talin, integ-rins are able to associate with the ECM, butare unable to connect to the cytoskeleton, lead-ing to muscle detachment at the integrin-actininterphase (Brown et al. 2002). Consistent withthis finding, mouse fibroblasts lacking bothtalin-1 and talin-2 are able to adhere, but notto link integrins to the cytoskeleton (Priddleet al. 1998; Zhang et al. 2008). This functionrequires the talin rod domain, which containsbinding sites for F-actin and vinculin

indicating that talin provides an essential linkageto the cytoskeleton independent of its ability toactivate integrins. Kindlins do not bind actindirectly, but connect to the actin cytoskele-ton via a migfilin-filamin interaction as wellas through the IPP complex (Montanez et al.2008). Platelets lacking kindlin-3 expressionare unable to organize their cytoskeleton orto establish stable lamellipodia (Moser et al.2008), which probably contributes to theirinability to spread and form a crosslinked clot(see Fig. 3). Loss of kindlin-2 impairs actinorganization and FA formation in endodermcells, which together with defective integrinactivation leads to early embryonic lethality.Interestingly, the rudimentary FAs in kindlin-2-deficient cells do not contain ILK, suggestingthat kindlin acts as a nucleator of FA formation(Montanez et al. 2008).

Despite the central role of talin and kindlinin outside-in signaling, they are not sufficient toestablish a stable integrin-actin connection. Animportant scaffold also required for this func-tion is the IPP complex, which is a constituentof at least b1 and b3 integrin-containing adhe-sion sites. The core component of this complexis ILK, a pseudokinase that is also capable ofbinding these integrins directly, at least in vitro(Wickstrom et al. 2010). Whether the bindingis actually relevant for the function of this com-plex, or whether it occurs in vivo, has not beenconclusively shown. The other two membersof this complex are pinch and parvin. Pinch,which interacts with the amino-terminalankyrin repeats of ILK, is a family of proteinscontaining only LIM domains and consistingof two members, pinch-1 and pinch-2 (Tuet al. 1999, 2001; Chiswell et al. 2008). The par-vins, which interact with the carboxy-terminalkinase-homology domain of ILK, are a familyof CH-domain-containing proteins with threemembers; the ubiquitously expressed a-parvin(also known as actopaxin or CH-ILKBP), b-parvin (also known as affixin), which is highlyexpressed in heart and skeletal muscle, and g-parvin, which is restricted to the hematopoieticsystem (Nikolopoulos and Turner 2000; Olskiet al. 2001; Tu et al. 2001; Yamaji et al. 2001;Chu et al. 2006). The IPP complex is thought

Genetic Analyses of Integrin Signaling

Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116 11

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 12: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

to be preassembled in the cytoplasm and is sub-sequently recruited to integrin adhesions in anILK-dependent manner (Zhang et al. 2002).The stability of each individual component de-pends on the assembly of the complex, as deple-tion of ILK or pinch leads to a decrease in theprotein levels of the other two complex mem-bers (Fukuda et al. 2003; Li et al. 2005).

The central biological function of the IPPcomplex is the regulation of the cytoskeletondownstream of integrins, although a numberof additional functions for this complex aswell as its individual components have beenassigned by in vitro studies. The importanceof these proteins in the integrin-actin connec-tion is highlighted by deletion studies in mice,where deletion of ILK or pinch-1 results in peri-implantation lethality caused by severe defectsin F-actin organization at adhesion sites, lead-ing to a failure in epiblast polarization (Sakaiet al. 2003; Li et al. 2005). The role of parvinsis more complex because of the presence ofthree structurally very similar isoforms. Micelacking b- or g-parvin show no obvious pheno-types, whereas a-parvin-null mice survive upto embryonic day 14.5 and die because ofdefects in cardiovascular development (Chuet al. 2006; Montanez et al. 2009; I Thievessenand R Fassler, unpubl.). Thea/b-parvin doubleknock-out results in early embryonic lethality,suggesting that parvins can compensate foreach other during early development (E. Mon-tanez and R. Fassler, unpubl.). Caenorhabditiselegans expresses only single orthologs for pinchand parvin, and is thus a suitable model organ-ism for a more straightforward analysis of theIPP complex. Studies in C. elegans have shownthat ILK (PAT-4), pinch (UNC-97), and parvin(PAT-6) colocalize with b integrin (PAT-3) atmuscle attachment sites, where a robust con-nection of cells to the cytoskeleton is requiredduring muscle contraction. Deletion of b in-tegrin or any member of the IPP complex leadsto detachment of muscles from the body walland embryonic lethality (Mackinnon et al.2002; Lin et al. 2003; Norman et al. 2007).

On the cellular level, ILK-deficiency leadsto compromised cell adhesion, spreadingand migration. This is because of defective

recruitment of the cytoskeleton to adhesionsites, resulting in defects in FA maturation andcytoskeletal remodeling. The defective mat-uration of ILK-deficient FAs into fibrillaradhesions subsequently leads to impaireddeposition of the FN matrix (Sakai et al. 2003;Stanchi et al. 2009). The precise molecularmechanism by which ILK regulates the cyto-skeleton is not clear. Parvins are capable ofinteracting with F-actin through their two CHdomains, but as these domains also regulatethe interaction of parvins with ILK and paxillin,they might not be available for actin binding.Therefore, it is likely that the IPP complexrequires additional downstream partners forthe regulation of the actin cytoskeleton, andthe identification of these proteins is a centralfocus of future research. It is, however, clearthat the presence of parvin in the IPP complexis critical for the regulation of the actin cyto-skeleton. The absence of a-parvin in micecauses impaired migration of vascular smoothmuscle cells (vSMC) toward developing vesselsresulting in defective stabilization of the vascu-lature and subsequent dilation of vessels, forma-tion of microaneurysms and vessel rupture. Themigration defect is caused by aberrant actomyo-sin contractility (Montanez et al. 2009), and canbe phenocopied by vSMC-specific ablation ofILK (Kogata et al. 2009). However, this functionseems to be restricted to certain cell types, sug-gesting that although the central role of theIPP complex in integrin outside-in signaling isubiquitous, the precise molecular mechanismsmight be cell type-specific to accommodatethe specialized needs of various cell and tissuetypes.

GENETIC DISEASES OF CELL–MATRIXINTERACTIONS

Aberrant cell–matrix interactions are involvedin a large number of pathological conditionssuch as cancer and various inflammatory dis-eases. Altered function of this signaling axis ishowever rarely the actual cause of these disor-ders. In contrast, recent studies have identifieda panel of genetic diseases whose etiology canbe pinpointed to a mutation in a component

S.A. Wickstrom et al.

12 Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 13: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

of the adhesive machinery. The high degree ofconservation in the components of this pathwayhas allowed the generation of mouse models forthese diseases that can be analyzed to generatemechanistic insights and therapeutic modalitiesfor clinical applications.

Integrin Activation Diseases

As discussed earlier, leukocyte and plateletadhesion are cellular events in which integrinaffinity regulation is of key importance. Inher-ited human diseases with defects in these proc-esses have been described more than 25 yearsago, but only recently, after the discovery ofthe central role of talin and kindlin in integrinactivation, the causes for these diseases havebeen identified.

There are currently three distinct syndromesthat together constitute the leukocyte adhesiondeficiency (LAD) family of diseases. They affectdistinct phases of the leukocyte adhesion cas-cade and therefore cause symptoms of variableseverity. LAD I, a disease that affects severalhundreds of patients worldwide, results fromimpaired firm adhesion of leukocytes, leadingto recurrent severe infections and impairedwound healing. This disease is caused by a rangeof mutations in the b2 integrin (ITGB2) gene,including deletions, truncations, substitutions,frame shifts, and intronic mutations, resultingin loss of protein expression or expression ofa truncated protein. LAD III (also known asLAD I/variant), which has been more recentlydescribed, is characterized by similar symptomsas LAD I. Interestingly, these patients also sufferfrom a bleeding tendency, indicating that addi-tional b subunits are involved. Indeed, thesepatients show defects in b1, b2, and b3 integrinactivation (Kuijpers et al. 1997; Alon andEtzioni 2003). In contrast to LAD I, no muta-tions in integrin genes have been identified,and the cause of this disease remained unknownuntil the role of kindlin in integrin activationwas discovered. Subsequently, genetic sequenc-ing of LAD III patients has revealed mutationsin the kindlin 3 (FRMT3) gene in all patientstested so far, leading to expression of a trun-cated protein, or reduction or loss of protein

expression (Mory et al. 2008; Kuijpers et al.2009; Malinin et al. 2009; Svensson et al.2009). Transfection of patient’s lymphocyteswith wild-type kindlin-3 was shown to restoreintegrin activation (Malinin et al. 2009; Svens-son et al. 2009). In addition, kindlin-3-defi-cient mice recapitulate all symptoms of LADIII, firmly identifying defective kindlin-3 asthe cause of LAD III (Moser et al. 2008; Moseret al. 2009a). In this respect, reconstitution ofkindlin-3 expression provides a possible thera-peutic modality for this disease. Interestingly,the naturally occurring mutations have alsoprovided information on the structure-func-tion aspects of kindlin-3. A homozygous stopcodon identified in three patients occurs distalto the PTB-containing integrin-binding subdo-main of kindlin-3, suggesting that the carboxylterminus of kindlin is required for its functionin integrin activation (Mory et al. 2008). A pointmutation leading to a truncation in the middleof the PH-domain was further shown to impairmembrane localization of kindlin-3 and therebyblock both lymphocyte adhesion and migra-tion, whereas a point mutation in the F2 sub-domain inhibited only the migration of thesecells, demonstrating that the functions of kind-lin in inside-out and outside-in signaling can beuncoupled (McDowall et al. 2010).

The bleeding tendency of LAD III patientsclosely resembles that of Glanzmann’s throm-basthenia (GT), a rare, autosomal recessivebleeding disorder caused by mutations leadingto quantitative or qualitative defects in plateletaIIbb3 integrin. The genetic background isvery heterogeneous: more than 100 differentmutations in either the aIIb (ITGA2B) or b3integrin (ITGB3) genes have been reported sofar (Kannan and Saxena 2009). Genetic ablationof the b3 integrin in mice almost fully reca-pitulates the human disease, including gastroin-testinal and cutaneous hemorrhage, increasedbleeding time, reduced platelet aggregation,and clot retraction (Hodivala-Dilke et al.1999). However, the heterogeneity in themutation spectrum of the integrins found inGT patients also applies to the symptoms; sib-lings sharing the same mutation can displaysymptoms of varying severity, suggesting that

Genetic Analyses of Integrin Signaling

Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116 13

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 14: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

polymorphisms in other genes involved inhemostasis could influence the phenotype. Inaddition, several GT patients lacking a mutationin the aIIbb3 integrin have been identified(Kannan and Saxena 2009). As the bleedingphenotype of the kindlin-3-deficient mice andthe LADIII patients closely resemble GT, it ispossible that altered function of this proteinmight play a role in this disease as well. How-ever, no polymorphisms in the kindlin-3 genein GT patients have been reported so far. Takentogether, integrin activation plays a central rolein immune function and hemostasis. Theprominent role of kindlin-3 in these processestogether with its restricted expression patternmakes it a promising candidate for anti-inflam-matory and antithrombotic therapies.

Adhesion Strengthening Diseases

Another “class” of diseases involving impairedcell–matrix interactions can be found affectingtissues that are subjected to high levels ofmechanical stress such as the skin and skeletalmuscle. The common denominator of thesedisorders, namely muscular dystrophies andskin-blistering diseases, is that the symptomsare caused by weakened interactions of cellswith their extracellular environment, leadingto tissue dysfunction and disruption. Anothercommon feature is that causative mutations fora single disorder can be found in either compo-nents of the ECM, their integrin receptors, oradhesion-strengthening scaffold proteins. Inthe case of congenital muscular dystrophies,although the dystrophin-dystroglycan-laminin/agrin axis is a common target (Yurchenco2010), other mutations affect the laminin-211/a7b1 integrin/plectin axis, leading to defectsin organization and migration of myoblasts,subsequent myofiber degeneration and progres-sive muscle weakness (for more detailed reviewsee Kanagawa and Toda 2006; Mendell et al.2006). Clinical features, cellular defects and theircorresponding causative mutations of musculardystrophies resulting from defective cell-ECMinteraction are summarized in Table 1.

The same principles are true for epidermol-ysis bullosa (EB), the prototype of human

skin-blistering diseases (Uitto 2009). EB is agroup of highly variable disorders characterizedby fragility of skin leading to blistering and ero-sions on mechanical trauma. Genetic studies ondifferent variants of EB have so far shown muta-tions in 12 distinct genes encoding structuralcomponents of the BM and adhesion proteins(Table 2). Interestingly, some of the EB patientsalso develop muscular dystrophy, underliningthe pathophysiological link between these twodiseases. As the histopathology of these diseasesis very complex, the identification of the causa-tive mutation has become a central diagnostictool of EB (Nagy and McGrath 2010). Geneticmouse models for the different EB variantsare available, and they have been used to workout therapeutic strategies for these diseases(Natsuga et al. 2010). As a result, gene delivery,ectopic protein replacement as well as cellulartherapies have already been tested on EBpatients (Uitto 2009).

CONCLUDING REMARKS

Two decades of genetic studies on cell-ECM in-teractions have illustrated the importance ofadhesion signaling in the development of mul-ticellular organisms as well as in disease. It isnow clear that these interactions not only pro-vide structural support and positional cues toguide morphogenesis, but also act as signalingplatforms to regulate cell fate in a highly tis-sue-specific manner. It is also evident that therelationship between ECM proteins and theirintegrin receptors is complex: Integrins trans-mit information from the ECM to the cell, butalso regulate the deposition and remodeling ofthe matrix itself. In addition, the ECM func-tions as a reservoir for growth factors, whereasintegrins cross talk with growth factor signal-ing on various levels. Because of this complex-ity, mechanistic interpretations of the variousphenotypes caused by genetic ablation of indi-vidual components of the adhesion signalingmachinery have proven to be difficult. Thus, amajor challenge for the future is to understandwhich types of intracellular signals directly ema-nate from integrin adhesions, and how thesesignals are propagated. This requires generation

S.A. Wickstrom et al.

14 Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 15: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

Table 1. Selected mutations causing muscular dystrophy and their corresponding mouse models

Human disease Central clinical features Affected gene (protein) Cellular defect Mouse model Reference

Limb-Girdlemusculardystrophy

Early adulthood onsetproximal muscle weakness,nasal pattern of speech

MYOT (myotilin) Sarcomeric organization N/A –

Severe, early onset muscleweakness

SGCA (a sarcoglycan) Muscle membraneintegrity

KO (Duclos et al. 1998)SGCB (b sarcoglycan) KO (Durbeej et al. 2000)SGCG (g sarcoglycan) KO (Sasaoka et al. 2003)SGCD (d sarcoglycan) KO (Coral-Vazquez et al. 1999)

Late onset muscle weaknessand atrophy,cardiomyopathy

TTN (titin) Sarcomere contraction/relaxation

N/A –

Relatively mild muscleweakness, variable clinicalfeatures

TCAP (telethonin) Sarcomere stability N/A –

Congenitalmusculardystrophy

Early onset with variablesymptoms, hypotonia,torticollis

ITGA7 (integrin a7) Impaired musclecell–matrix interactions

KO (Mayer et al. 1997)

Early onset hypotonia andmuscle weakness

COL6A1COL6A2COL6A3 (type VI

collagen)

Stability of extracellularmatrix

KOa (Bonaldo et al. 1998)

Severe, early onset muscleweakness, white matterhypodensity, mentalretardation

LAMA2 (laminin 211) Attachment, migrationand organization ofmyoblasts

Dy/Dy (naturally occurringmutant)

(Sunada et al. 1994)

Dy2j/Dy2j (naturallyoccurring mutant)

(Xu et al. 1994)

DyPas/DyPas (naturallyoccurring null allele)

(Besse et al. 2003)

Dy3K/Dy3K (KO) (Miyagoe et al. 1997)DyW/DyW (hypomorph) (Kuang et al. 1998)

Duchennemusculardystrophy

Severe, early onset muscleweakness with respiratory,orthopedic and cardiaccomplications

DMD (dystrophin) Stabilization of membraneduring contraction

Mdx (naturally occurring nullallele)

(Bulfield et al. 1984)

Mdx 2Cv, Mdx 3CvMdx 4Cv, Mdx 5Cvb

(Chapman et al. 1989)

Mdx 52 (deletion of exon 52) (Araki et al. 1997)aAblation of the col6a1 gene.bMutants recovered from ENU chemical mutagenesis screen.

Abbreviations: N/A, not analyzed; KO, knockout mouse; Dy, Dystrophin; mdx, X chromosome-linked muscular dystrophy.

Gen

eticA

nalyses

of

Integrin

Signalin

g

Cite

this

articleas

Cold

Sprin

gH

arbPersp

ectB

iol2011;3

:a005116

15

on July 23, 2021 - Published by C

old Spring H

arbor Laboratory Press

http://cshperspectives.cshlp.org/D

ownloaded from

Page 16: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

Table 2. Mutations causing epidermolysis bullosa (EB) and their corresponding mouse models

Human diseaseSite of tissueseparation Affected gene (protein) Mouse model Mouse skin phenotype Reference

Epidermolysis

bullosa simplex(EBS)

Intraepidermal PKP1 (plakophilin-1) N/A – –

DSP (desmoplakin) N/A – –KRT5 (keratin-5) KOa Severe blistering, loss of keratin

filaments(Peters et al. 2001)

KRT14 (keratin-14) KO Blistering, decreased keratinfilaments

(Lloyd et al. 1995)

K14-R131C KIa Severe blistering (Cao et al. 2001)K14-DCT TG Blistering (Vassar et al. 1991)

PLEC1 (plectin) KOa Blistering, reduced stability andnumber of hemidesmosomes

(Andra et al. 1997)

Conditional KO Blistering, skin fragility (Ackerl et al. 2007)Junctional

epidermolysisbullosa (JBS)

Lamina lucida ITGA6 (integrin a6) KOa Severe blistering, loss ofhemidesmosomes

(Georges-Labouesse et al. 1996a)

ITGB4 (integrin b4) KOa Severe blistering, loss ofhemidesmosomes

(Dowling et al. 1996; van der Neutet al. 1996)

ITGB4-DCT KIa Severe blistering, loss ofhemidesmosomes,hypoproliferation

(Murgia et al. 1998)

Conditional KO Blistering, loss of hemidesmosomes (Raymond et al. 2005)LAMA3 (laminin-332) KOa Severe blistering, abnormal

hemidesmosomes, reduced cellsurvival

(Ryan et al. 1999)

LAMB3 (laminin-332) Spontaneous null allele Blistering, abnormalhemidesmosomes

(Kuster et al. 1997)

LAMC2 (laminin-332) KOa Blistering, rudimentary

hemidesmosomes, apoptosis

(Meng et al. 2003)

Spontaneoushypomorphic allele

Blistering, ulcerations,hyperkeratosis, rudimentaryhemidesmosomes

(Bubier et al. 2010)

Dystrophicepidermolysisbullosa (DBS)

Sub-lamina densa COL7A1 (type VII collagen) KOa Severe blistering, absence ofanchoring fibrils

(Heinonen et al. 1999)

COL7 hypomorph Severe blistering, decreasedanchoring fibrils

(Fritsch et al. 2008)

Kindler syndrome Primarily lamina

lucida

FERMT1 (kindlin-1) KOa Skin atrophy (Ussar et al. 2008)

aLeads to early postnatal lethality.

Abbreviations used: N/A, not analyzed; KO, knock-out mouse; K14-R131C, Arginine-131 to Cysteine substitution in the Keratin-14 gene; KI, knock in mouse; K14-DCT, carboxy-

terminal truncation of Keratin-14; ITGB4-DCT, deletion of the entire cytoplamsic domain of integrin b4; TG, transgenic mouse.

S.A.W

ickstrom

etal.

16C

iteth

isarticle

asC

old

Sprin

gH

arbPersp

ectB

iol2011;3

:a005116

on July 23, 2021 - Published by C

old Spring H

arbor Laboratory Press

http://cshperspectives.cshlp.org/D

ownloaded from

Page 17: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

of more sophisticated mouse models, such asknock-in mice carrying point mutations thatonly partially disrupt protein function togetherwith reporter mice for specific signaling events,transcriptional changes, or changes in protein-protein interactions or conformations. In addi-tion, a large majority of the cell-biological anal-yses on integrin signaling are still performedon rigid 2D-surfaces, which very poorly reca-pitulate the more compliant and complex 3Denvironment of tissues. As our understandingof the biophysical properties of the ECM in tis-sues steadily increases, a major goal of futureresearch is to translate this knowledge into gen-eration of more relevant in vitro models for cellbiological studies of integrin signaling.

ACKNOWLEDGMENTS

The authors apologize to all those whose workcould not be cited because of space restrictionsand for not always citing all the primary litera-ture. The authors would like to thank MaxIglesias for artwork. The work of the Wickstromand Fassler laboratories are funded by the MaxPlanck Society.

REFERENCES

Ackerl R, Walko G, Fuchs P, Fischer I, Schmuth M, Wiche G.2007. Conditional targeting of plectin in prenatal andadult mouse stratified epithelia causes keratinocyte fragil-ity and lesional epidermal barrier defects. J Cell Sci 120:2435–2443.

Alon R, Etzioni A. 2003. LAD-III, a novel group of leukocyteintegrin activation deficiencies. Trends Immunol 24:561–566.

Alt A, Ohba M, Li L, Gartsbein M, Belanger A, Denning MF,Kuroki T, Yuspa SH, Tennenbaum T. 2001. Protein kinaseCd-mediated phosphorylation of a6b4 is associatedwith reduced integrin localization to the hemidesmo-some and decreased keratinocyte attachment. CancerRes 61: 4591–4598.

Andra K, Lassmann H, Bittner R, Shorny S, Fassler R, PropstF, Wiche G. 1997. Targeted inactivation of plectin revealsessential function in maintaining the integrity of skin,muscle, and heart cytoarchitecture. Genes Dev 11:3143–3156.

Araki E, Nakamura K, Nakao K, Kameya S, Kobayashi O,Nonaka I, Kobayashi T, Katsuki M. 1997. Targeted dis-ruption of exon 52 in the mouse dystrophin gene inducedmuscle degeneration similar to that observed in Duch-enne muscular dystrophy. Biochem Biophys Res Commun238: 492–497.

Astrof S, Crowley D, Hynes RO. 2007. Multiple cardiovascu-lar defects caused by the absence of alternatively splicedsegments of fibronectin. Dev Biol 311: 11–24.

Bader BL, Rayburn H, Crowley D, Hynes RO. 1998. Exten-sive vasculogenesis, angiogenesis, and organogenesisprecede lethality in mice lacking all av integrins. Cell95: 507–519.

Bauer M, Brakebusch C, Coisne C, Sixt M, Wekerle H,Engelhardt B, Fassler R. 2009. Beta1 integrins differen-tially control extravasation of inflammatory cell subsetsinto the CNS during autoimmunity. Proc Natl Acad Sci106: 1920–1925.

Bazigou E, Xie S, Chen C, Weston A, Miura N, Sorokin L,Adams R, Muro AF, Sheppard D, Makinen T. 2009.Integrin-a9 is required for fibronectin matrix assemblyduring lymphatic valve morphogenesis. Dev Cell 17:175–186.

Bertotti A, Comoglio PM, Trusolino L. 2006. b4 integrinactivates a Shp2-Src signaling pathway that sustainsHGF-induced anchorage-independent growth. J CellBiol 175: 993–1003.

Besse S, Allamand V, Vilquin JT, Li Z, Poirier C, Vignier N,Hori H, Guenet JL, Guicheney P. 2003. Spontaneousmuscular dystrophy caused by a retrotransposal insertionin the mouse laminina2 chain gene. Neuromuscul Disord13: 216–222.

Bonaldo P, Braghetta P, Zanetti M, Piccolo S, Volpin D, Bres-san GM. 1998. Collagen VI deficiency induces early onsetmyopathy in the mouse: An animal model for Bethlemmyopathy. Hum Mol Genet 7: 2135–2140.

Brakebusch C, Grose R, Quondamatteo F, Ramirez A, Jor-cano JL, Pirro A, Svensson M, Herken R, Sasaki T, TimplR, et al. 2000. Skin and hair follicle integrity is cruciallydependent on b1 integrin expression on keratinocytes.EMBO J 19: 3990–4003.

Brown NH, Gregory SL, Rickoll WL, Fessler LI, Prout M,White RA, Fristrom JW. 2002. Talin is essential for integ-rin function in Drosophila. Dev Cell 3: 569–579.

Bubier JA, Sproule TJ, Alley LM, Webb CM, Fine JD,Roopenian DC, Sundberg JP. 2010. A mouse model ofgeneralized non-Herlitz junctional epidermolysis bul-losa. J Invest Dermatol 130: 1819–1828.

Bulfield G, Siller WG, Wight PA, Moore KJ. 1984. X chromo-some-linked muscular dystrophy (mdx) in the mouse.Proc Natl Acad Sci 81: 1189–1192.

Campbell ID, Humphries MJ. 2011. Integrin structure, acti-vation, and interactions. Cold Spring Harb Perspect Bioldoi: 10.1101/cshperspect.a004994.

Cao T, Longley MA, Wang XJ, Roop DR. 2001. An induciblemouse model for epidermolysis bullosa simplex: Impli-cations for gene therapy. J Cell Biol 152: 651–656.

Chapman VM, Miller DR, Armstrong D, Caskey CT. 1989.Recovery of induced mutations for X chromosome-linked muscular dystrophy in mice. Proc Natl Acad Sci86: 1292–1296.

Chiswell BP, Zhang R, Murphy JW, Boggon TJ, CalderwoodDA. 2008. The structural basis of integrin-linked kinase-PINCH interactions. Proc Natl Acad Sci 105: 20677–20682.

Chu H, Thievessen I, Sixt M, Lammermann T, Waisman A,Braun A, Noegel AA, Fassler R. 2006. gamma-Parvin is

Genetic Analyses of Integrin Signaling

Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116 17

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 18: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

dispensable for hematopoiesis, leukocyte trafficking, andT-cell-dependent antibody response. Mol Cell Biol 26:1817–1825.

Coral-Vazquez R, Cohn RD, Moore SA, Hill JA, Weiss RM,Davisson RL, Straub V, Barresi R, Bansal D, Hrstka RF,et al. 1999. Disruption of the sarcoglycan-sarcospancomplex in vascular smooth muscle: A novel mechanismfor cardiomyopathy and muscular dystrophy. Cell 98:465–474.

Critchley DR, Gingras AR. 2008. Talin at a glance. J Cell Sci121: 1345–1347.

de Pereda JM, Lillo MP, Sonnenberg A. 2009. Structuralbasis of the interaction between integrin a6b4 and plec-tin at the hemidesmosomes. EMBO J 28: 1180–1190.

DiPersio CM, van der Neut R, Georges-Labouesse E, Kreid-berg JA, Sonnenberg A, Hynes RO. 2000. aa3b1 anda6b4 integrin receptors for laminin-5 are not essentialfor epidermal morphogenesis and homeostasis duringskin development. J Cell Sci 113: 3051–3062.

Dowling J, Yu QC, Fuchs E. 1996.ba4 integrin is required forhemidesmosome formation, cell adhesion and cell sur-vival. J Cell Biol 134: 559–572.

Duclos F, Straub V, Moore SA, Venzke DP, Hrstka RF, CrosbieRH, Durbeej M, Lebakken CS, Ettinger AJ, van derMeulen J, et al. 1998. Progressive muscular dystrophy ina-sarcoglycan-deficient mice. J Cell Biol 142: 1461–1471.

Durbeej M, Cohn RD, Hrstka RF, Moore SA, Allamand V,Davidson BL, Williamson RA, Campbell KP. 2000. Dis-ruption of the b-sarcoglycan gene reveals pathogeneticcomplexity of limb-girdle muscular dystrophy type 2E.Mol Cell 5: 141–151.

Erickson AC, Couchman JR. 2000. Still more complexityin mammalian basement membranes. J Histochem Cyto-chem 48: 1291–1306.

Fritsch A, Loeckermann S, Kern JS, Braun A, Bosl MR, BleyTA, Schumann H, von Elverfeldt D, Paul D, Erlacher M,et al. 2008. A hypomorphic mouse model of dystrophicepidermolysis bullosa reveals mechanisms of diseaseand response to fibroblast therapy. J Clin Invest 118:1669–1679.

Fukuda T, Chen K, Shi X, Wu C. 2003. PINCH-1 is an obli-gate partner of integrin-linked kinase (ILK) functioningin cell shape modulation, motility, and survival. J BiolChem 278: 51324–51333.

Garcia-Alvarez B, de Pereda JM, Calderwood DA, Ulmer TS,Critchley D, Campbell ID, Ginsberg MH, Liddington RC.2003. Structural determinants of integrin recognition bytalin. Mol Cell 11: 49–58.

Garmy-Susini B, Avraamides CJ, Schmid MC, Foubert P,Ellies LG, Barnes L, Feral C, Papayannopoulou T, LowyA, Blair SL, et al. 2010. Integrin a4b1 signaling isrequired for lymphangiogenesis and tumor metastasis.Cancer Res 70: 3042–3051.

Geerts D, Fontao L, Nievers MG, Schaapveld RQ, Purkis PE,Wheeler GN, Lane EB, Leigh IM, Sonnenberg A. 1999.Binding of integrina6b4 to plectin prevents plectin asso-ciation with F-actin but does not interfere with inter-mediate filament binding. J Cell Biol 147: 417–434.

Geiger B, Yamada KM. 2011. Molecular architecture andfunction of matrix adhesions. Cold Spring Harb PerspectBiol doi: 10.1101/cshperspect.a005033.

George EL, Baldwin HS, Hynes RO. 1997. Fibronectins areessential for heart and blood vessel morphogenesis butare dispensable for initial specification of precursor cells.Blood 90: 3073–3081.

George EL, Georges-Labouesse EN, Patel-King RS, RayburnH, Hynes RO. 1993. Defects in mesoderm, neural tubeand vascular development in mouse embryos lackingfibronectin. Development 119: 1079–1091.

Georges-Labouesse EN, George EL, Rayburn H, Hynes RO.1996b. Mesodermal development in mouse embryosmutant for fibronectin. Dev Dyn 207: 145–156.

Georges-Labouesse E, Messaddeq N, Yehia G, Cadalbert L,Dierich A, Le Meur M. 1996a. Absence of integrin a6leads to epidermolysis bullosa and neonatal death inmice. Nat Genet 13: 370–373.

Geuijen CA, Sonnenberg A. 2002. Dynamics of the alpha6-beta4 integrin in keratinocytes. Mol Biol Cell 13: 3845–3858.

Giancotti FG. 2007. Targeting integrin b4 for cancerand anti-angiogenic therapy. Trends Pharmacol Sci 28:506–511.

Guan JL, Hynes RO. 1990. Lymphoid cells recognize analternatively spliced segment of fibronectin via the integ-rin receptor a4 b1. Cell 60: 53–61.

Guo L, Degenstein L, Dowling J, Yu QC, Wollmann R,Perman B, Fuchs E. 1995. Gene targeting of BPAG1:Abnormalities in mechanical strength and cell migrationin stratified epithelia and neurologic degeneration. Cell81: 233–243.

Guo W, Pylayeva Y, Pepe A, Yoshioka T, Muller WJ, Inghi-rami G, Giancotti FG. 2006. b4 integrin amplifiesErbB2 signaling to promote mammary tumorigenesis.Cell 126: 489–502.

Heinonen S, Mannikko M, Klement JF, Whitaker-MenezesD, Murphy GF, Uitto J. 1999. Targeted inactivation ofthe type VII collagen gene (Col7a1) in mice results insevere blistering phenotype: A model for recessive dystro-phic epidermolysis bullosa. J Cell Sci 112: 3641–3648.

Hodivala-Dilke KM, McHugh KP, Tsakiris DA, Rayburn H,Crowley D, Ullman-Cullere M, Ross FP, Coller BS, Teitel-baum S, Hynes RO. 1999. b4-integrin-deficient miceare a model for Glanzmann thrombasthenia showingplacental defects and reduced survival. J Clin Invest 103:229–238.

Huang XZ, Wu JF, Ferrando R, Lee JH, Wang YL, Farese RVJr, Sheppard D. 2000. Fatal bilateral chylothorax in micelacking the integrin a9b1. Mol Cell Biol 20: 5208–5215.

Humphries JD, Byron A, Humphries MJ. 2006. Integrinligands at a glance. J Cell Sci 119: 3901–3903.

Hynes RO. 2002. Integrins: Bidirectional, allosteric signal-ing machines. Cell 110: 673–687.

Hynes RO. 2009. The extracellular matrix: Not just prettyfibrils. Science 326: 1216–1219.

Ivaska J, Heino J. 2010. Interplay between cell adhesion andgrowth factor receptors: From the plasma membrane tothe endosomes. Cell Tissue Res 339: 111–120.

Kanagawa M, Toda T. 2006. The genetic and molecular basisof muscular dystrophy: Roles of cell-matrix linkage in thepathogenesis. J Hum Genet 51: 915–926.

Kannan M, Saxena R. 2009. Glanzmann’s thrombasthenia:An overview. Clin Appl Thromb Hemost 15: 152–165.

S.A. Wickstrom et al.

18 Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 19: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

Kennel SJ, Foote LJ, Lankford PK. 1981. Analysis of surfaceproteins of mouse lung carcinomas using monoclonalantibodies. Cancer Res 41: 3465–3470.

Kogata N, Tribe RM, Fassler R, Way M, Adams RH. 2009.Integrin-linked kinase controls vascular wall formationby negatively regulating Rho/ROCK-mediated vascularsmooth muscle cell contraction. Genes Dev 23: 2278–2283.

Koster J, Kuikman I, Kreft M, Sonnenberg A. 2001. Two dif-ferent mutations in the cytoplasmic domain of the integ-rin b4 subunit in nonlethal forms of epidermolysisbullosa prevent interaction of b4 with plectin. J InvestDermatol 117: 1405–1411.

Koster J, van Wilpe S, Kuikman I, Litjens SH, Sonnenberg A.2004. Role of binding of plectin to the integrin b4 sub-unit in the assembly of hemidesmosomes. Mol Biol Cell15: 1211–1223.

Kuang W, Xu H, Vachon PH, Liu L, Loechel F, Wewer UM,Engvall E. 1998. Merosin-deficient congenital musculardystrophy. Partial genetic correction in two mouse mod-els. J Clin Invest 102: 844–852.

Kuijpers TW, Van Lier RA, Hamann D, de Boer M, Thung LY,Weening RS, Verhoeven AJ, Roos D. 1997. Leukocyteadhesion deficiency type 1 (LAD-1)/variant. A novelimmunodeficiency syndrome characterized by dysfunc-tional beta2 integrins. J Clin Invest 100: 1725–1733.

Kuijpers TW, van de Vijver E, Weterman MA, de Boer M,Tool AT, van den Berg TK, Moser M, Jakobs ME, SeegerK, Sanal O, et al. 2009. LAD-1/variant syndrome iscaused by mutations in FERMT3. Blood 113: 4740–4746.

Kuster JE, Guarnieri MH, Ault JG, Flaherty L, Swiatek PJ.1997. IAP insertion in the murine LamB3 gene resultsin junctional epidermolysis bullosa. Mamm Genome 8:673–681.

Kwee L, Baldwin HS, Shen HM, Stewart CL, Buck C, BuckCA, Labow MA. 1995. Defective development of theembryonic and extraembryonic circulatory systems invascular cell adhesion molecule (VCAM-1) deficientmice. Development 121: 489–503.

Legate KR, Fassler R. 2009. Mechanisms that regulate adap-tor binding to {b}-integrin cytoplasmic tails. J Cell Sci122: 187–198.

Legate KR, Wickstrom SA, Fassler R. 2009. Genetic and cellbiological analysis of integrin outside-in signaling. Genes& Dev 23: 397–418.

Leiss M, Beckmann K, Giros A, Costell M, Fassler R. 2008.The role of integrin binding sites in fibronectin matrixassembly in vivo. Curr Opin Cell Biol 20: 502–507.

Li S, Bordoy R, Stanchi F, Moser M, Braun A, Kudlacek O,Wewer UM, Yurchenco PD, Fassler R. 2005. PINCH1 reg-ulates cell-matrix and cell-cell adhesions, cell polarity andcell survival during the peri-implantation stage. J Cell Sci118: 2913–2921.

Lin X, Qadota H, Moerman DG, Williams BD. 2003. C. ele-gans PAT-6/actopaxin plays a critical role in the assemblyof integrin adhesion complexes in vivo. Curr Biol 13:922–932.

Litjens SH, de Pereda JM, Sonnenberg A. 2006. Currentinsights into the formation and breakdown of hemides-mosomes. Trends Cell Biol 16: 376–383.

Litjens SH, Koster J, Kuikman I, van Wilpe S, de Pereda JM,Sonnenberg A. 2003. Specificity of binding of the plectinactin-binding domain to b4 integrin. Mol Biol Cell 14:4039–4050.

Lloyd C, Yu QC, Cheng J, Turksen K, Degenstein L, HuttonE, Fuchs E. 1995. The basal keratin network of stratifiedsquamous epithelia: Defining K15 function in theabsence of K14. J Cell Biol 129: 1329–1344.

Mackinnon AC, Qadota H, Norman KR, Moerman DG,Williams BD. 2002. C. elegans PAT-4/ILK functions asan adaptor protein within integrin adhesion complexes.Curr Biol 12: 787–797.

Malinin NL, Zhang L, Choi J, Ciocea A, Razorenova O, MaYQ, Podrez EA, Tosi M, Lennon DP, Caplan AI, et al.2009. A point mutation in KINDLIN3 ablates activationof three integrin subfamilies in humans. Nat Med 15:313–318.

Mao Y, Schwarzbauer JE. 2005. Fibronectin fibrillogenesis,a cell-mediated matrix assembly process. Matrix Biol24: 389–399.

Margadant C, Frijns E, Wilhelmsen K, Sonnenberg A. 2008.Regulation of hemidesmosome disassembly by growthfactor receptors. Curr Opin Cell Biol 20: 589–596.

Mariotti A, Kedeshian PA, Dans M, Curatola AM, Gagnoux-Palacios L, Giancotti FG. 2001. EGF-R signaling throughFyn kinase disrupts the function of integrin a6b4 athemidesmosomes: Role in epithelial cell migration andcarcinoma invasion. J Cell Biol 155: 447–458.

Mayer U, Saher G, Fassler R, Bornemann A, Echtermeyer F,von der Mark H, Miosge N, Poschl E, von der Mark K.1997. Absence of integrin a7 causes a novel form ofmuscular dystrophy. Nat Genet 17: 318–323.

McDowall A, Svensson L, Stanley P, Patzak I, Chakravarty P,Howarth K, Sabnis H, Briones M, Hogg N. 2010. Twomutations in the KINDLIN3 gene of a new leukocyteadhesion deficiency-III patient reveal distinct effects onleukocyte function in vitro. Blood 115: 4834–4842.

Mendell JR, Boue DR, Martin PT. 2006. The congenitalmuscular dystrophies: Recent advances and molecularinsights. Pediatr Dev Pathol 9: 427–443.

Meng X, Klement JF, Leperi DA, Birk DE, Sasaki T, Timpl R,Uitto J, Pulkkinen L. 2003. Targeted inactivation of mu-rine laminin g2-chain gene recapitulates human junc-tional epidermolysis bullosa. J Invest Dermatol 121:720–731.

Miner JH. 2008. Laminins and their roles in mammals.Microsc Res Tech 71: 349–356.

Miner JH, Li C, Mudd JL, Go G, Sutherland AE. 2004. Com-positional and structural requirements for laminin andbasement membranes during mouse embryo implanta-tion and gastrulation. Development 131: 2247–2256.

Miyagoe Y, Hanaoka K, Nonaka I, Hayasaka M, NabeshimaY, Arahata K, Nabeshima Y, Takeda S. 1997. Laminina2 chain-null mutant mice by targeted disruption ofthe Lama2 gene: A new model of merosin (laminin2)-deficient congenital muscular dystrophy. FEBS Lett415: 33–39.

Montanez E, Ussar S, Schifferer M, Bosl M, Zent R, MoserM, Fassler R. 2008. Kindlin-2 controls bidirectional sig-naling of integrins. Genes Dev 22: 1325–1330.

Genetic Analyses of Integrin Signaling

Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116 19

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 20: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

Montanez E, Wickstrom SA, Altstatter J, Chu H, Fassler R.2009. aa-parvin controls vascular mural cell recruitmentto vessel wall by regulating RhoA/ROCK signalling.EMBO J 28: 3132–3144.

Mory A, Feigelson SW, Yarali N, Kilic SS, Bayhan GI,Gershoni-Baruch R, Etzioni A, Alon R. 2008. Kindlin-3:A new gene involved in the pathogenesis of LAD-III.Blood 112: 2591.

Moser M, Bauer M, Schmid S, Ruppert R, Schmidt S, SixtM, Wang HV, Sperandio M, Fassler R. 2009a. Kindlin-3is required for b2 integrin-mediated leukocyte adhesionto endothelial cells. Nat Med 15: 300–305.

Moser M, Legate KR, Zent R, Fassler R. 2009b. The tail ofintegrins, talin, and kindlins. Science 324: 895–899.

Moser M, Nieswandt B, Ussar S, Pozgajova M, Fassler R.2008. Kindlin-3 is essential for integrin activation andplatelet aggregation. Nat Med 14: 325–330.

Murgia C, Blaikie P, Kim N, Dans M, Petrie HT, GiancottiFG. 1998. Cell cycle and adhesion defects in mice carryinga targeted deletion of the integrin beta4 cytoplasmicdomain. EMBO J 17: 3940–3951.

Nagy N, McGrath JA. 2010. Blistering skin diseases: A bridgebetween dermatopathology and molecular biology.Histopathology 56 91–99.

Natsuga K, Shinkuma S, Nishie W, Shimizu H. 2010. Animalmodels of epidermolysis bullosa. Dermatol Clin 28:137–142.

Nieswandt B, Moser M, Pleines I, Varga-Szabo D, MonkleyS, Critchley D, Fassler R. 2007. Loss of talin1 in plateletsabrogates integrin activation, platelet aggregation, andthrombus formation in vitro and in vivo. J Exp Med204: 3113–3118.

Nievers MG, Kuikman I, Geerts D, Leigh IM, Sonnenberg A.2000. Formation of hemidesmosome-like structures inthe absence of ligand binding by the (a)6(b)4 integrinrequires binding of HD1/plectin to the cytoplasmicdomain of the (b)4 integrin subunit. J Cell Sci 113:963–973.

Nievers MG, Schaapveld RQ, Oomen LC, Fontao L, GeertsD, Sonnenberg A. 1998. Ligand-independent role of theb4 integrin subunit in the formation of hemidesmo-somes. J Cell Sci 111: 1659–1672.

Nikolopoulos SN, Turner CE. 2000. Actopaxin, a new focaladhesion protein that binds paxillin LD motifs and actinand regulates cell adhesion. J Cell Biol 151: 1435–1448.

Nikolopoulos SN, Blaikie P, Yoshioka T, Guo W, GiancottiFG. 2004. Integrin beta4 signaling promotes tumorangiogenesis. Cancer Cell 6: 471–483.

Nikolopoulos SN, Blaikie P, Yoshioka T, Guo W, Puri C,Tacchetti C, Giancotti FG. 2005. Targeted deletion ofthe integrin b4 signaling domain suppresses laminin-5-dependent nuclear entry of mitogen-activated proteinkinases and NF-kB, causing defects in epidermal growthand migration. Mol Cell Biol 25: 6090–6102.

Nishie W, Sawamura D, Goto M, Ito K, Shibaki A, McMillanJR, Sakai K, Nakamura H, Olasz E, Yancey KB, et al. 2007.Humanization of autoantigen. Nat Med 13: 378–383.

Norman KR, Cordes S, Qadota H, Rahmani P, MoermanDG. 2007. UNC-97/PINCH is involved in the assemblyof integrin cell adhesion complexes in Caenorhabditiselegans body wall muscle. Dev Biol 309: 45–55.

Olski TM, Noegel AA, Korenbaum E. 2001. Parvin, a 42 kDafocal adhesion protein, related to the a-actinin super-family. J Cell Sci 114: 525–538.

Pankov R, Yamada KM. 2002. Fibronectin at a glance. J CellSci 115: 3861–3863.

Peters B, Kirfel J, Bussow H, Vidal M, Magin TM. 2001.Complete cytolysis and neonatal lethality in keratin 5knockout mice reveal its fundamental role in skin integ-rity and in epidermolysis bullosa simplex. Mol Biol Cell12: 1775–1789.

Petrich BG, Marchese P, Ruggeri ZM, Spiess S, Weichert RA,Ye F, Tiedt R, Skoda RC, Monkley SJ, Critchley DR, et al.2007. Talin is required for integrin-mediated plateletfunction in hemostasis and thrombosis. J Exp Med 204:3103–3111.

Priddle H, Hemmings L, Monkley S, Woods A, Patel B,Sutton D, Dunn GA, Zicha D, Critchley DR. 1998. Dis-ruption of the talin gene compromises focal adhesionassembly in undifferentiated but not differentiatedembryonic stem cells. J Cell Biol 142: 1121–1133.

Rabinovitz I, Toker A, Mercurio AM. 1999. Protein kinaseC-dependent mobilization of the a6b4 integrin fromhemidesmosomes and its association with actin-richcell protrusions drive the chemotactic migration of carci-noma cells. J Cell Biol 146: 1147–1160.

Rabinovitz I, Tsomo L, Mercurio AM. 2004. Protein kinaseC-aphosphorylation of specific serines in the connectingsegment of the b4 integrin regulates the dynamics of typeII hemidesmosomes. Mol Cell Biol 24: 4351–4360.

Raymond K, Kreft M, Janssen H, Calafat J, Sonnenberg A.2005. Keratinocytes display normal proliferation, sur-vival and differentiation in conditional b4-integrinknockout mice. J Cell Sci 118: 1045–1060.

Rezniczek GA, de Pereda JM, Reipert S, Wiche G. 1998.Linking integrin a6b4-based cell adhesion to the inter-mediate filament cytoskeleton: Direct interaction be-tween the b4 subunit and plectin at multiple molecularsites. J Cell Biol 141: 209–225.

Ryan MC, Lee K, Miyashita Y, Carter WG. 1999. Targeteddisruption of the LAMA3 gene in mice reveals abnormal-ities in survival and late stage differentiation of epithelialcells. J Cell Biol 145: 1309–1323.

Sakai T, Li S, Docheva D, Grashoff C, Sakai K, Kostka G,Braun A, Pfeifer A, Yurchenco PD, Fassler R. 2003.Integrin-linked kinase (ILK) is required for polarizingthe epiblast, cell adhesion, and controlling actin accumu-lation. Genes & Dev 17: 926–940.

Santoro MM, Gaudino G, Marchisio PC. 2003. The MSPreceptor regulates a6b4 and a3b1 integrins via 14–3-3proteins in keratinocyte migration. Dev Cell 5: 257–271.

Sasaoka T, Imamura M, Araishi K, Noguchi S, Mizuno Y,Takagoshi N, Hama H, Wakabayashi-Takai E, Yoshi-moto-Matsuda Y, Nonaka I, et al. 2003. Pathological anal-ysis of muscle hypertrophy and degeneration in musculardystrophy in gamma-sarcoglycan-deficient mice. Neuro-muscul Disord 13: 193–206.

Schwarzbauer JE, DeSimone DW. 2011. Deciphering themechanisms of fibronectin matrix assembly. Cold SpringHarb Perspect Biol doi: 10.1101/cshperspect.a005041.

Shattil SJ, Kim C, Ginsberg MH. 2010. The final steps ofintegrin activation: the end game. Nat Rev Mol Cell Biol11: 288–300.

S.A. Wickstrom et al.

20 Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 21: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

Simonson WT, Franco SJ, Huttenlocher A. 2006. Talin1 reg-ulates TCR-mediated LFA-1 function. J Immunol 177:7707–7714.

Smyth N, Vatansever HS, Murray P, Meyer M, Frie C, Pauls-son M, Edgar D. 1999. Absence of basement membranesafter targeting the LAMC1 gene results in embryoniclethality due to failure of endoderm differentiation.J Cell Biol 144: 151–160.

Sonawane M, Carpio Y, Geisler R, Schwarz H, MaischeinHM, Nuesslein-Volhard C. 2005. Zebrafish penner/lethalgiant larvae 2 functions in hemidesmosome formation,maintenance of cellular morphology and growth regula-tion in the developing basal epidermis. Development 132:3255–3265.

Sonawane M, Martin-Maischein H, Schwarz H, Nusslein-Volhard C. 2009. Lgl2 and E-cadherin act antagonis-tically to regulate hemidesmosome formation duringepidermal development in zebrafish. Development 136:1231–1240.

Stanchi F, Grashoff C, Nguemeni Yonga CF, Grall D, FasslerR, Van Obberghen-Schilling E. 2009. Molecular dis-section of the ILK-PINCH-parvin triad reveals a fun-damental role for the ILK kinase domain in the latestages of focal-adhesion maturation. J Cell Sci 122:1800–1811.

Steinman L. 2009. A molecular trio in relapse and remissionin multiple sclerosis. Nat Rev Immunol 9: 440–447.

Sterk LM, Geuijen CA, Oomen LC, Calafat J, Janssen H,Sonnenberg A. 2000. The tetraspan molecule CD151, anovel constituent of hemidesmosomes, associates withthe integrin a6b4 and may regulate the spatial organiza-tion of hemidesmosomes. J Cell Biol 149: 969–982.

Sunada Y, Bernier SM, Kozak CA, Yamada Y, Campbell KP.1994. Deficiency of merosin in dystrophic dy mice andgenetic linkage of laminin M chain gene to dy locus.J Biol Chem 269: 13729–13732.

Svensson L, Howarth K, McDowall A, Patzak I, Evans R,Ussar S, Moser M, Metin A, Fried M, Tomlinson I,et al. 2009. Leukocyte adhesion deficiency-III is causedby mutations in KINDLIN3 affecting integrin activation.Nat Med 15: 306–312.

Tadokoro S, Shattil SJ, Eto K, Tai V, Liddington RC, dePereda JM, Ginsberg MH, Calderwood DA. 2003. Talinbinding to integrin b tails: a final common step in integ-rin activation. Science 302: 103–106.

Takahashi S, Leiss M, Moser M, Ohashi T, Kitao T, Heck-mann D, Pfeifer A, Kessler H, Takagi J, Erickson HP,et al. 2007. The RGD motif in fibronectin is essentialfor development but dispensable for fibril assembly.J Cell Biol 178: 167–178.

Tan MH, Sun Z, Opitz SL, Schmidt TE, Peters JH, GeorgeEL. 2004. Deletion of the alternatively spliced fibronectinEIIIA domain in mice reduces atherosclerosis. Blood 104:11–18.

Tanentzapf G, Brown NH. 2006. An interaction betweenintegrin and the talin FERM domain mediates integrinactivation but not linkage to the cytoskeleton. Nat CellBiol 8: 601–606.

Tanentzapf G, Martin-Bermudo MD, Hicks MS, BrownNH. 2006. Multiple factors contribute to integrin-talininteractions in vivo. J Cell Sci 119: 1632–1644.

Tasanen K, Tunggal L, Chometon G, Bruckner-Tuderman L,Aumailley M. 2004. Keratinocytes from patients lackingcollagen XVII display a migratory phenotype. Am JPath 164: 2027–2038.

Trusolino L, Bertotti A, Comoglio PM. 2001. A signalingadapter function for a6b4 integrin in the control ofHGF-dependent invasive growth. Cell 107: 643–654.

Tsuruta D, Hopkinson SB, Jones JC. 2003. Hemidesmosomeprotein dynamics in live epithelial cells. Cell Motil Cyto-skeleton 54: 122–134.

Tsuruta D, Kobayashi H, Imanishi H, Sugawara K, Ishii M,Jones JC. 2008. Laminin-332-integrin interaction: A tar-get for cancer therapy? Curr Med Chem 15: 1968–1975.

Tu Y, Huang Y, Zhang Y, Hua Y, Wu C. 2001. A new focaladhesion protein that interacts with integrin-linked kin-ase and regulates cell adhesion and spreading. J Cell Biol153: 585–598.

Tu Y, Li F, Goicoechea S, Wu C. 1999. The LIM-only proteinPINCH directly interacts with integrin-linked kinase andis recruited to integrin-rich sites in spreading cells. MolCell Biol 19: 2425–2434.

Uematsu J, Nishizawa Y, Sonnenberg A, Owaribe K. 1994.Demonstration of type II hemidesmosomes in a mam-mary gland epithelial cell line, BMGE-H. J Biochem115: 469–476.

Uitto J. 2009. Progress in heritable skin diseases: Transla-tional implications of mutation analysis and prospectsof molecular therapies. Acta Derm Venereol 89: 228–235.

Underwood RA, Carter WG, Usui ML, Olerud JE. 2009.Ultrastructural localization of integrin subunits b4 anda3 within the migrating epithelial tongue of in vivohuman wounds. J Histochem Cytochem 57: 123–142.

Ussar S, Moser M, Widmaier M, Rognoni E, Harrer C,Genzel-Boroviczeny O, Fassler R. 2008. Loss of Kindlin-1causes skin atrophy and lethal neonatal intestinal epithe-lial dysfunction. PLoS Genet 4: e1000289

Ussar S, Wang HV, Linder S, Fassler R, Moser M. 2006. Thekindlins: Subcellular localization and expression duringmurine development. Exp Cell Res 312: 3142–3151.

Vajkoczy P, Laschinger M, Engelhardt B. 2001. Alpha4-integrin-VCAM-1 binding mediates G protein-inde-pendent capture of encephalitogenic T cell blasts toCNS white matter microvessels. J Clin Invest 108:557–565.

van der Neut R., Krimpenfort P, Calafat J, Niessen CM, Son-nenberg A. 1996. Epithelial detachment due to absence ofhemidesmosomes in integrinb4 null mice. Nat Genet 13:366–369.

Vassar R, Coulombe PA, Degenstein L, Albers K, Fuchs E.1991. Mutant keratin expression in transgenic micecauses marked abnormalities resembling a humangenetic skin disease. Cell 64: 365–380.

Wayner EA, Garcia-Pardo A, Humphries MJ, McDonald JA,Carter WG. 1989. Identification and characterization ofthe T lymphocyte adhesion receptor for an alternativecell attachment domain (CS-1) in plasma fibronectin.J Cell Biol 109: 1321–1330.

Weiss P, Ferris W. 1954. Electron-microscopic study of thetexture of the basement membrane of larval amphibianskin. Proc Natl Acad Sci 40: 528–540.

Genetic Analyses of Integrin Signaling

Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116 21

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 22: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

Wickstrom SA, Lange A, Montanez E, Fassler R. 2010. TheILK/PINCH/parvin complex: the kinase is dead, longlive the pseudokinase! EMBO J 29: 281–291.

Wilhelmsen K, Litjens SH, Kuikman I, Margadant C, vanRheenen J, Sonnenberg A. 2007. Serine phosphorylationof the integrin b4 subunit is necessary for epidermalgrowth factor receptor induced hemidesmosome disrup-tion. Mol Biol Cell 18: 3512–3522.

Wright MD, Geary SM, Fitter S, Moseley GW, Lau LM,Sheng KC, Apostolopoulos V, Stanley EG, Jackson DE,Ashman LK. 2004. Characterization of mice lacking thetetraspanin superfamily member CD151. Mol Cell Biol24: 5978–5988.

Xu H, Wu XR, Wewer UM, Engvall E. 1994. Murine muscu-lar dystrophy caused by a mutation in the laminin a2(Lama2) gene. Nat Genet 8: 297–302.

Yamaji S, Suzuki A, Sugiyama Y, Koide Y, Yoshida M,Kanamori H, Mohri H, Ohno S, Ishigatsubo Y. 2001. Anovel integrin-linked kinase-binding protein, affixin, isinvolved in the early stage of cell-substrate interaction.J Cell Biol 153: 1251–1264.

Yang JT, Rayburn H, Hynes RO. 1993. Embryonic mesoder-mal defects in a5 integrin-deficient mice. Development119: 1093–1105.

Yang JT, Rayburn H, Hynes RO. 1995. Cell adhesion eventsmediated by a4 integrins are essential in placental andcardiac development. Development 121: 549–560.

Yang JT, Bader BL, Kreidberg JA, Ullman-Cullere M,Trevithick JE, Hynes RO. 1999. Overlapping andindependent functions of fibronectin receptor integrinsin early mesodermal development. Dev Biol 215:264–277.

Yednock TA, Cannon C, Fritz LC, Sanchez-Madrid F, Stein-man L, Karin N. 1992. Prevention of experimental auto-immune encephalomyelitis by antibodies against a4 b1integrin. Nature 356: 63–66.

Yurchenco PD. 2010. Basement membranes: cell scaffold-ings and signaling platforms. Cold Spring Harb PerspectBiol doi: 10.1101/cshperspect.a004911.

Zaidel-Bar R, Itzkovitz S, Ma’ayan A, Iyengar R, Geiger B.2007. Functional atlas of the integrin adhesome. NatCell Biol 9: 858–867.

Zhang Y, Chen K, Tu Y, Velyvis A, Yang Y, Qin J, Wu C. 2002.Assembly of the PINCH-ILK-CH-ILKBP complexprecedes and is essential for localization of each com-ponent to cell-matrix adhesion sites. J Cell Sci 115:4777–4786.

S.A. Wickstrom et al.

22 Cite this article as Cold Spring Harb Perspect Biol 2011;3:a005116

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 23: Genetic Analyses of Integrin Signalingcshperspectives.cshlp.org/content/3/2/a005116.full.pdf · 2011. 2. 2. · Genetic Analyses of Integrin Signaling Sara A. Wickstro¨m1, Korana

December 30, 20102011; doi: 10.1101/cshperspect.a005116 originally published onlineCold Spring Harb Perspect Biol 

 Sara A. Wickström, Korana Radovanac and Reinhard Fässler Genetic Analyses of Integrin Signaling

Subject Collection Extracellular Matrix Biology

and VertebratesMechanisms Conserved between Invertebrates Extracellular Matrix in Development: Insights from

Nicholas H. Brown

in Development and DiseaseExtracellular Matrix Degradation and Remodeling

Pengfei Lu, Ken Takai, Valerie M. Weaver, et al.

ThrombosisExtracellular Matrix Proteins in Hemostasis and

Wolfgang Bergmeier and Richard O. HynesExtracellular Matrix Constituents and Functions

An Inventory of−−Overview of the Matrisome

Richard O. Hynes and Alexandra NabaThe Thrombospondins

Josephine C. Adams and Jack LawlerIntegrins in Cell Migration

Anna Huttenlocher and Alan Rick Horwitz

Integrins, and the Extracellular Matrix Signaling Pathways,βCross Talk among TGF-

John S. Munger and Dean SheppardFunctionsFibronectins, Their Fibrillogenesis, and In Vivo

Jean E. Schwarzbauer and Douglas W. DeSimoneHeparan Sulfate Proteoglycans

Jeffrey D. EskoStephane Sarrazin, William C. Lamanna and System

Extracellular Matrix: Functions in the Nervous

MüllerClaudia S. Barros, Santos J. Franco and Ulrich

The Collagen FamilySylvie Ricard-Blum Adhesions

Molecular Architecture and Function of Matrix

Benjamin Geiger and Kenneth M. Yamada

ModulationTenascins and the Importance of Adhesion

Ruth Chiquet-Ehrismann and Richard P. Tuckerand Diseased SkinCell-Extracellular Matrix Interactions in Normal

Fiona M. Watt and Hironobu FujiwaraIntegrin Structure, Activation, and Interactions

Iain D. Campbell and Martin J. HumphriesGenetic Analyses of Integrin Signaling

Reinhard FässlerSara A. Wickström, Korana Radovanac and

http://cshperspectives.cshlp.org/cgi/collection/ For additional articles in this collection, see

Copyright © 2011 Cold Spring Harbor Laboratory Press; all rights reserved

on July 23, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from