U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Madame Curie Bioscience Database [Internet]. Austin (TX): Landes Bioscience; 2000-2013.

Cover of Madame Curie Bioscience Database

Madame Curie Bioscience Database [Internet].

Show details

The Role of Integrins in Cell Migration

.

Author Information and Affiliations

Introduction

Migration and Motility are essential components of the behavioral repertoire of a cell. During embryogenesis cells move in sheets or loosely attached populations to create complex tissues. In the adult, cell motility is crucial to maintain immunity, or repair damaged tissues. Uncontrolled cell adhesion and increased motility can result in pathological situations such as tumor cell dissemination and the formation of metastasis.

Cell migration occurs in response to chemokine or growth factor signals that are converted into cell shape changes, essentially mediated by structural changes of the actin cytoskeleton. Cell displacement is observed when the structural changes and forces created within the actin cytoskeleton are mechanically linked to the surrounding extracellular scaffold. This mechanical link needs to be highly regulated in order to allow the formation of new attachment sites at the cell front and the controlled dissociation of adhesion sites at the cell rear.

The understanding of the mechanisms that govern cell migration is therefore critically coupled to the question of how cellular receptors interact with their extracellular environment. Hence, members of the family of integrins have moved into the focus of attention. Integrins are heterodimeric membrane spanning receptors that play crucial roles in cell adhesion and migration. 1 This chapter will discuss general as well as specific aspects of how integrins are involved in the orchestration of the migration of individual cells.

Evolutionary Aspects of Integrin Dependent Cell Migration

When analyzing the appearance of integrin receptors during evolution, it is notable that unicellular highly mobile organisms, such as Dictyostelium, do not possess integrins. Apparently, the different types of substrates encountered in the soil, the natural habitat of these organisms, require weaker and less specific means of adhesions as provided by integrins. However, studies on the movement and chemotactic migration of amoeba demonstrated the fundamental role of actin polymerization and anterior to posterior polarization.2-4 Moreover, since classical integrin adaptor molecules such as talin can be found in Dictyostelium,5,6 we have to propose that integrins evolved in a cellular environment in which the entire cytoskeletal machinery that drives cell migration was already in place. Based on the appearance of integrins in metazoan cells, they might have played a role in the development of early multicellular organisms such as sponges and jellyfish.7-10 Hence both the firm adhesion to extracellular matrix as well as the morphogenetic movements that occur during embryogenesis were supported by this new class of cell surface receptors.11 Therefore, integrins have evolved to perform critical functions in cell adhesion as well as cell motility in complex multicellular organisms.

The Role of Integrins in Cell Migration in Fly's and Worm's

Genetically well characterized biological systems such as C. elegans and Drosophila confirm the essential roles played by integrins in cell to matrix adhesion, such as the formation of the attachment sites for muscle cells or the adhesion between the dorsal and ventral wing epithelium (for reviews see refs. 12,13). Similar to the structural aspects of cell adhesion, the mechanisms that regulate cell migration have also been analyzed in these animals. Classical models of cell migration include the border cell migration, midgut migration and the development of the tracheal system in Drosophila, and neuronal and gonadal distal tip cell migration in C. elegans. The nonbiased genetic approach established roles for growth factor signaling during the development of the tracheal system,14 cell-cell adhesion molecules in border cell migration15 and integrins in midgut migration.16 In addition, genetic analysis has established critical roles for the cytoplasmic integrin adaptor proteins talin and UNC-112 in cell adhesion and migration.17- 19 The importance of these additional molecular components reveal that the adhesive and signaling functions mediated by integrins during cell migration are embedded in a complex network of protein-protein interactions, intracellular signaling and mechanisms controlling polarity and intracellular protein transport.20-22 In order to better understand the roles of integrins in migration, mutations that specifically affect cell migration but not cell adhesion are of particular interest. In C. elegans, the tyrosine to phenylalanine mutation of the two phylogenetically conserved tyrosine residues in the cytoplasmic tail of the unique β-integrin subunit (βpat-3), blocks the migration of gonadal distal tip cells but does not affect muscle function.23 Thus, it is tempting to speculate that the role of integrins in cell migration involves the specific activation of intracellular signaling pathways as well as the dynamic interactions with molecules of the extracellular matrix. In fact, the challenge consists in understanding the mechanisms that allow a cell to reversibly engage integrins into cell-matrix adhesions sites and exploit them for adhesion as well as migration.

Definition of Cell Migration

Traditionally, fibroblasts have been used to study cell adhesion and migration.24,25 Therefore the prototype of cellular motion has been established with these cells migrating on two-dimensional surfaces. As the study on other moving cells, such as leukocytes or epithelial cells has advanced, some of the hallmarks of cell adhesions need to be modified. Moreover, the role of the respective architecture of the 3-dimensional environment has to be taken into account in order to compare the movement of keratinocytes on top of a flat basement matrix with the slow tunneling of contractile fibroblasts within a three-dimensional collagen network.26-29 Hence it is important to define the cellular processes that occur during cell migration in such a way that they are applicable for many different cell types within different tissue environments.

Cell migration is initiated by the development of cellular extensions such as filopodia and lamellipodia that define the front of migrating cells.30 Within these cellular protrusions, small punctuate integrin-containing contacts form at sites of physical interaction with extracellular matrix components. These sites are referred to as “focal complexes”. Focal complexes anchor the molecular scaffold formed by the polymerized actin cytoskeleton to the extracellular matrix. Once the actin cytoskeleton is linked to the extracellular matrix, new actin molecules that are added to the actin cytoskeletal scaffold at the tips or leading edges of filopodia and lamellipodia respectively push the plasma membrane forward.31 In turn, the newly added extensions are stabilized by a new set of focal complexes formed in front of the previous ones.

The second phase of cell migration is initiated when focal complexes mature into “focal contacts”. This maturation is dependent on intracellular or extracellular mechanical tension and localized actin polymerization and results in the compaction of the actin cytoskeleton and integrin receptors, creating a robust link between intracellular actin fibers (stress fibers) and the extracellular matrix.32-34 In rapidly migrating leukocytes or fish keratinocytes, the formation of stress fibers and focal contacts is not observed, suggesting that the development of stress fibers and focal contacts is associated with cell anchorage rather than mobility.35-38 During migration of fibroblasts, it has been observed that integrins and trailing parts of the cell remain on the substrate while the main cell body is moving forward.39 Interestingly, the substrate affinity of the trailing cell rear, and hence the maximal speed of migration, depends on the concentration of extracellular ligands. This observation led to the important conclusion that maximal migration speeds of fibroblasts are obtained on low concentrations of ligands.40,41 Hence the destabilization of stress fibers and their associated focal contacts may represent a target for intracellular signaling pathways that increase the speed of cell migration (see below).

Cell migration however cannot occur unless integrin dependent adhesion sites are dispersed at the rear of migrating cells. Before dispersal, focal contacts often demonstrate an apparent sliding in the direction of migration. This sliding is associated with a continuous reduction of focal contact size.26,27,34,42 We recently proposed that the apparent sliding of focal contacts is the result of a polarized remodeling, created by the dispersal of integrin receptors at the distal and the recruitment of new integrin receptors at the proximal edge of sliding focal contacts respectively.26,27

In summary, cells of different origins migrate in an integrin dependent manner, all according to the same principles, involving (i) the formation of extensions at the cell front, (ii) integrin dependent focal complex formation, (iii) maturation into and plasticity of focal contacts and (iv) the controlled sliding and dispersal of focal contacts at the cell rear. In this chapter, we will use the terms focal complexes and focal contacts as defined above. The term “focal adhesions” will be used to refer to not further specified integrin dependent cell to matrix adhesion sites. We will now analyze the different roles of integrins applying this concept of cell migration.

Differential Roles of Specific Integrins in Cell Migration

24 different types of integrin heterodimers have been identified in vertebrates.1 Due to their evolutionary and functional relationship, they can be grouped into sub-families performing different mechanical or signaling tasks, such as the recognition of a subset of extracellular ligands, being involved in the assembly of extracellular matrix or playing a regulatory role in cell migration.8 The genetic inactivation of individual integrin chains and the resulting pathologies has established critical and distinct roles for each type of integrin, identifying roles in cell migration for most of them.

The β1-Integrin Family

The β1-integrin family is characterized by a total of 12 different heterodimers that all share a common β1-integrin subunit. While the β1-integrin chain provides the critical link to the actin cytoskeleton in each of these heterodimers, the associated α-chains determine ligand specificity, cellular localization and function in adhesion and/or migration.

α1 β1, α2 β1, α10 β1 and α11 β1 integrins are receptors for collagen and support cell migration through collagen rich extracellular matrix. It has been demonstrated that the expression of α1 and α2 chains are increased upon VEGF secretion in response to injury and are required for lymphangiogenesis in vivo.43 A similar increase in α1 expression and associated collagen remodeling is induced by growth factors such as TGFβ and PDGF.44 In addition, an increase in α1 expression, in response to chemokines such as MCP-1 and IL-8, is induced in infiltrating lymphocytes.45 In this context, it has been demonstrated that the infiltration and remodeling of collagen matrices is facilitated by the recruitment of the metalloproteinase MMP1 by the I-domain of the α2 integrin subunit.46 α10β1 expression is limited to chondrocytes and provides binding to cartilage specific type II collagen.47 Its genetic inactivation results in the dysfunction of growth plate chondrocytes.48 α11β1 integrin is expressed in a subset of mesenchymal tissues and modulates PDGF dependent chemotaxis on collagen matrixes.49,50

α3 β1, α6 β1 and α7 β1 integrins bind to laminin, an important constituent of basement membranes. Although these integrins bind to various members of the laminin family of extracellular ligands, their differential expression in brain, epithelial and muscle tissues determines different roles in adhesion and migration.

The genetic inactivation of α3 β1 revealed a phenotype of weak skin blistering, kidney tubule defects, reduced branching during lung morphogenesis and a lamination defect in the neurocortex.51 Of interest is the aberrant neuronal migration that occurs in the absence of α3-integrin and which results in altered actin cytoskeleton dynamics at the leading edge of migrating neurons.52 During the migration of neurons from the ventricular zone through the cortex towards the pia mater, α3β1 integrin expression is tightly regulated. In scrambler mice the timely expression of α3-integrin is altered, resulting in prolonged neuronal migration and positioning defects. It has been demonstrated that the phosphorylation on tyrosines of disabled- 1, the protein mutated in scrambler mice, induces the expression of α3 integrins in neurons. In response to the expression of α3β1 integrin at the cell surface, neurons migrating along radial glial cells, detach from these cells to adhere to the extracellular matrix protein laminin. The timely disconnection from the glial cell surface results in the correct positioning of the neuron within the neurocortex.53

In contrast to migration defects observed upon the loss of α3β1 integrin, its over-expression has been associated with increased formation of metastases of small cell lung carcinomas and colon carcinomas.54-56

α6 β1 integrin is of importance for the migration and adhesion of epithelial cells on laminin containing basement membranes. The deletion of the α6-integrin chain results in a severe skin-blistering defect.57 However, this defect of epithelial adhesion is attributed to the absence of the α6β4 integrin heterodimer.58,59 It demonstrates the unique role of the β4-integrin subunit to link this particular laminin binding integrin to the intermediate filament system and not to the actin cytoskeleton.60,61 Because of the crucial role of α6β4, the importance of the α6β1 integrin in morphogenesis and maintenance of epithelial tissues has not been clearly demonstrated. However, indirect evidence for the importance of the actin cytoskeleton-linked β1 integrin has been provided by the phenotype of the Kindler syndrome, in which affected individuals exhibit fragile skin.62 The gene mutated in this syndrome is a β1-integrin adaptor protein named kindlerin belonging to the family of mig-2 related adaptor proteins, sharing homologies with UNC112 from C. elegans.63 Kindlerin expression is increased in breast carcinomas upon TGFβ treatment and localizes to focal adhesions in keratinocytes.64

In other systems such as the developing cerebellar and cerebral cortex, α6β1 integrin and more generally the β1 integrins are required to define the structural entity of the tissue. In the absence of α6-integrins, neuronal migration along the surface of radial glial cells is not affected, suggesting that glial dependent neuronal migration is mediated by cell-cell adhesion receptors. However, in the absence of α6-integrin, the situation is similar to that of α3-integrin deficient mice, in which migrating neuronal precursors do not stop their migration along radial glial cells and form neuronal outgrowths beyond the pia mater.65,66 In contrast, β1-integrins are required to physically anchor the endfeet of the radial glial cells, allowing these cells to span the entire neurocortex and thereby forming the neuronal migration pathways and contributing to the stabilization of the brain architecture.67

Another situation in which the regulation of the cell surface expression of α6β1 integrin is important for cell migration has been described in extravasating neutrophils. PECAM-1 dependent binding between neutrophils and endothelial cells results in the up-regulation of α6β1 integrins on the surface of neutrophils, which is critical for their transmigration through the perivascular basement membrane.68

Furthermore, in migrating cranial neural crest cells, the cycling of the α6β1 integrin between the cell surface and an intracellular pool, is a mechanism that promotes neural crest cell motility.69

α7 β1 integrin is found expressed in muscle cells where its deletion results in a particular form of congenital muscular dystrophy.70-72 Despite the important function for the maintenance of muscles, the specific function of α7β1 or their specific splice variants in myoblast migration during embryogenesis, the fusion of myotubes or muscle regeneration is not well studied.73-76

α4 β1 and α9 β1 are dual specific integrins that recognize fibronectin at sites distinct from the major RGD containing cell binding site and VCAM-1 exposed on endothelial cells.77,78 Although several members of the integrin family are involved in cell migration, the two structurally related α4β1 and α9β1 integrins appear to specifically enhance cell migration. For example, the initiation of α4β1 integrin expression in the neural crest coincides with the onset of neural crest migration. When the function of this integrin is blocked, both the number of neural crest cells and the distance of migration are reduced.79,80

During inflammation, leukocyte migration across the endothelium is facilitated by the interaction of α4β1 with VCAM-1. The pathological importance of this mechanism for the progression of auto-immune diseases such as multiple sclerosis or Crohn's disease has been recently demonstrated by the successful treatment of these conditions by a humanized anti-α4β1 integrin antibody (for a review see ref. 81).

It has been determined that the cytoplasmic tail of the α4-integrin subunit is responsible for the increase in migration of leukocytes.82 Integrin α4β1 induced cell migration depends on the interaction of the α4 cytoplasmic tail with the focal adhesion adaptor protein paxillin.83 However, in polarized migrating cells, the fraction of α4β1 integrin that localizes to the leading edge is not associated with paxillin. In fact, in this cellular location, paxillin binding to α4 is prevented by phosphorylation of a critical serine residue in the α4 cytoplasmic tail. Upon dephosphorylation of α4 integrin, paxillin can bind to α4β1 integrins, which results in their colocalization in focal complexes at the cell front and in focal contacts at the cell rear.84 The reversible binding of paxillin to α4β1 integrins is critically required for cell migration, since cell migration is completely inhibited in cells transfected with an α4-integrin/paxillin fusion protein.84 These data suggest that the recruitment of paxillin to α4β1 integrins has to follow a strict chronological order, required for the assembly of focal complexes at the cell front.85 Furthermore, it has been demonstrated that paxillin localized to focal contacts located at the cell rear is able to prevent the formation of new lamellipodia by the recruitment of an ARF-GAP, and thus contributes to the stabilization of the anterior to posterior cell polarity required for efficient migration.86 Another critical intracellular target of the α4 integrin /paxillin complex that could be responsible for the stabilization of cell polarity during migration, represents the focal adhesion kinase/proline-rich tyrosine kinase-2. The α4-integrin/paxillin dependent activation of this kinase stimulates αLβ2 dependent trans-endothelial migration.87

α9 β1 integrin also interacts with paxillin, but it lacks the regulatory phosphorylation site and enhances cell motility independent of paxillin binding.88 It was recently proposed that α9 dependent stimulation of cell migration is mediated by the specific recruitment of the spermidine/spermine N1-acetyltransferase to the α9 subunit.89 However, the issue concerning the role of α9β1 integrin in stimulating cell migration remains puzzling. The genetic deletion of α9 integrin results in a fatal bilateral chylothorax, probable linked to an abnormal development of the lymphatic system.90 Accordingly, it has recently been demonstrated that the lymphangiogenesis inducing growth factors VEGF-C and VEGF-D bind to α9β1 integrins.91

α5 β1 integrin is an RGD-peptide dependent receptor for fibronectin and has been studied in great detail. This integrin supports adhesion of migrating cells to fibronectin, but is also critically involved in the assembly of fibronectin containing extracellular fibrils.92,93 In fibroblasts cultured on a rigid 2-dimensional matrix, this integrin localizes in focal contacts at the cell periphery as well as to fibrillar adhesions located underneath the main cell body. Interestingly, an increase in the elasticity of the extracellular matrix reduces the phenotypic distinction between focal contacts and fibrillar adhesion.94 It has been recently demonstrated that the α5 integrin subunit localized in fibrillar adhesions undergoes a structural change that induces the high affinity binding of the α5β1 integrin to extracellular fibronectin. The firm mechanical coupling across the plasma membrane enables fibroblasts to synthesize fibronectin fibrils by the acto-myosin dependent pulling of α5β1 integrins along stress fibers.95

By using a GFP-tagged form of α5-integrin, it became possible to follow the clustering as well as the recycling of α5β1 integrins in migrating fibroblasts. Importantly, it was observed that α5β1 integrins were internalized at the cell rear and transported to the front.96 Recently the dynamics of α5β1 integrin in the plasma membrane of living cells was analyzed by image correlation microscopy.97 It was demonstrated that α5β1 integrins exists in submicroscopic clusters moving together with α-actinin. In addition, it was determined that α5β1 integrins are loosely clustered within focal complexes, but densely packed and highly structured in focal contacts, confirming observations previously made for αvβ3 integrins.34

α8 β1 plays a crucial role during early steps of kidney morphogenesis, affecting the growth and branching of the ureteric bud epithelium.98 Interestingly, α8β1 integrin is expressed in the metanephric mesenchyme surrounding the ureteric bud. Its major ligand nephronectin is expressed in the ureteric bud epithelium from where it is deposited in the surrounding basement membrane.99 Thus the role of α8β1 integrin in the migration or movement of the ureteric bud epithelium is indirect and may be linked to α8β1 integrin induced mesenchyme to epithelial signaling. Interestingly, it has recently been demonstrated that α8β1 is able to bind in an RGD dependent manner to a latent form of TGFβ.100 Nephronectin might therefore be able to compete for the binding of TGFβ making it available for morphogenetic events in the tissue. Although α8β1 may not be directly involved in the stimulation of cell migration in a cell autonomous manner, its expression on vascular smooth muscle cells and myofibroblasts indicates a role in injury related scarring of the myocardium.101

αv β1 integrin has specificity for RGD containing extracellular ligands. Because the αv subunit can bind several different β-integrin sububits, the αv family of integrins will be discussed below.

The β2 Integrin Family

The expression of integrins belonging to this family is the hallmark of cells of the immune system. The β2-integrin subunit forms various heterodimers with I-like containing α-integrin subunits ( αL β2, αMβ2, αX β2, αDβ2), enabling their interactions with counter receptors of the IgG-domain family on the surface of activated endothelial cells or multiple ligands of the extracellular matrix.102 These diverse interactions allow firm adhesion of chemokine-stimulated leukocytes to endothelial cells, followed by diapedesis and chemotaxis through damaged or infected tissues. Compared to other migrating cells, leukocytes demonstrate the most impressive motility. In contrast to fibroblasts, leukocytes lack stress fibers and focal contacts.

The best studied leukocyte integrin, the αL β2 integrin is switched to its high affinity form and recruited to the front of migrating leukocytes by the specific interaction with RAPL.103,104 In order to be able to bind αLβ2 integrins, RAPL is stimulated by Rap1, a member of the small GTPase family of intracellular regulators that plays an important role in the activation of integrins.105,106 It has been demonstrated that RAPL binds to the juxtamembrane region of the αL subunit, in such a way that it may provoke the unclasping of the transmembrane and cytoplasmic domains of the αL and β2 integrin subunits.107- 109 Thus RAPL has a dual role consisting in the activation of αLβ2 and in its recruitment to the leading edge of migrating leukocytes, preparing αLβ2 integrin for binding to ligands expressed on the endothelial cell surface.

Another unique feature of β2 integrins is their continuous recycling from the trailing uropod (cell rear of migrating leukocytes), to the cell front. This process requires the activity of myosin light chain kinase and intracellular Ca2+ transients. If either of these two intracellular signaling pathways are blocked, rear detachment of the uropod is prevented, leading to the arrest of leukocyte migration.110-113 The ability of β2-integrins to undergo rapid recycling has been linked to a juxtamembrane localized tyrosine dependent YXXØ internalization motif.114

Interestingly, this motif is also present in the β7-integrin subunit, which like β2-integrin is expressed exclusively in cells of the hemaotopoietic system.114 The β7-integrin subunit pairs with the α4 and αE integrin chains forming α4β7 and αEβ7 integrins. α4β7 is critical for the morphogenesis of the gut-associated lymphatic system,115 while αEβ7 regulates immuno-surveillance of the skin.116 The β7-integrins are also involved in CD8+ T-cell mediated graft rejection and consist of a promising therapeutic target to enhance the success of tissue transplantation.117,118

The αMβ2 integrin is expressed in monocytes, granulocytes, macrophages and natural killer cells and has been implicated in diverse responses of these cells, including phagocytosis, cell-mediated killing and chemotaxis. These complex responses depend on the capacity of αMβ2 integrin to mediate leukocyte adhesion and migration, playing a central role in inflammation. 102 In contrast to αLβ2 integrin, αMβ2 and also αX β2 integrins recognize a large set of structurally unrelated ligands, including fibrinogen, complement fragment iC3b, ICAM-1, Facor X, JAM's and denatured proteins.119,120 Recently it has been suggested that the reason for this large spectrum of ligands is the unusual high affinity of the αMβ2 and αXβ2 for carboxyl groups. Moreover, since both αMβ2 and αXβ2 integrins are colocalized with the receptor for the urokinase-type plasminogen activator to the leading edge of migrating cells,121,122 it has been proposed that proteolytic degradation of extracellular matrix molecules such as fibrinogen creates new extracellular ligands specifically recognized by these two integrins.120 Using these integrins as sensors for degraded extracellular matrix components would provide leukocytes with an additional tool to detect tissue damage or microbial invasion.

αDβ2 integrin is expressed on eosinophiles, monocyte/macrophages and neutrophils and is an alternative receptor for VCAM-1. Compared to the other β2-integrin family member its role in leukocyte migration is less studied. In an experimental model of spinal cord injury, it has been reported that the injection of monoclonal antibodies directed against αDβ2 reduces the number of lesion-induced accumulation of macrophages or neutrophils by approximately 50%.123

Besides αLβ2 recycling and RAPL dependent targeting to the cell front, it is not clear whether other β2 integrin subunit specific features are responsible for the high migratory capacity of leukocytes. When comparing the cytoplasmic domain of β2 integrins with that of other β-integrin chains, it is notable that the two NPXY motifs found in all other β-subunits exist as NPXF motifs in β2. Interestingly, when both NPXY to NPXF mutations are introduced into the cytoplasmic domain of the β3-integrin gene, the so mutated platelet integrin αIIbβ3 (see below) is no longer able to mediate blood clot retraction, leading to prolonged bleeding.124 The same β-integrin mutation, when expressed in the context of the αvβ3 integrin, blocks shear stress induced reinforcement of cell adhesions125 and inhibits βPat-3 dependent migration of gonadal distal tip cells in C. elegans.23 Thus the capacity for fast cell migration and reliable transmigration of leukocytes at sites of chemokine release might be directly linked to the inability of β2 integrins to sense the shear forces constantly generated by the flowing blood.

The αv-Integrin Family

The αv-integrin subunit pairs with a series of different β-subunits ( αv β1, αv β3, αv β5, αv β6, αv β8) forming heterodimers with specificities for RGD peptide containing ligands. Among these different heterodimers, αv β3 integrin is the most studied in association with cell migration. Notably, the expression of αvβ3 integrin is up-regulated in endothelial cells that undergo angiogenesis, and is over-expressed in mammary carcinomas and melanomas.126-128 In melanomas, the expression of αvβ3 is associated with the formation of metastasis and the protection against cell death in 3-D collagen matrixes.128,129 Moreover, an increase in the formation of metastasis has been induced by the expression of a constitutively activated form of the αvβ3 integrin in breast carcinoma cells.130 Due to its increased expression on endothelial cells during tumor induced angiogenesis, αvβ3 integrin has been the center of attention and the focus of the development of ligand mimetic small ligands, that are able to inhibit angiogenesis. 131,132 Interestingly one of these small soluble ligands, cyclic RGD peptide, is able to induce caspase-3 dependent apoptosis.133 As a paradox however, the knockout of β3-integrin and the double knockout together with β5 integrin increases tumor-induced angiogenesis.134 Hence αvβ3 integrin plays an essential role in the regulating of cell migration, in ways that have not been understood yet.

Based on the pathological importance of the αv-integrin family, it was surprising to see only minor defects in β3-integrin knockout animals. Similarly, the knockout of the β5-integrin demonstrated a slight increase in osteoclast maturation and activity, while other β5-integrin specific functions such as wound healing and adenovirus susceptibility were normal.135,136 This may suggest that the β3 or β5-integrin can compensate for each other. However, several findings demonstrate that they nevertheless play different roles during cell migration. While αvβ3 expressed in CS-1 hamster melanoma cells was able to induce adhesion, spreading and migration, αvβ5 provided only adhesion. Interestingly, cell migration was induced when αvβ5 expressing cells were stimulated with cytokines. This difference in behavior was attributed to the extracellular domain of these different β-integrins.137 While both integrins bind to vitronectin, only αvβ5 integrin is able to rapidly internalize and degrade soluble vitronectin.138 It was recently demonstrated that the constitutive internalization of αvβ5 is of physiological importance. β5-integrin knockout mice develop an age related blindness that is due to a lack of phagocytic activity of the retinal pigment epithelium, normally involved in the uptake of defective photoreceptor rods that are shed by retinal ganglion neurons.139

The inactivation of the gene coding for the β6-integrin subunit results in the chronic inflammation of skin and lungs. Accordingly, keratinocyte migration on fibronectin and vitronectin is reduced.140 The lung phenotype can be rescued by the constitutive expression of a β6-integrin transgene in bronchial and alveolar epithelial cells.141 Since β6-integrin expression is increased during the repair of the airway epithelium, the epithelial expression of αvβ6 integrin could be crucial for the proliferation, adhesion and or migration of epithelial cells.

The expression of β8-integrin is elevated in the CNS compared to other organs. In the CNS, it is localized to dendritic spines and post-synaptic densities, however, a critical role in neuronal function has so far not been demonstrated.142

Although the size of the αv-integrin family is considerable, the inactivation of the αv-integrin gene does not result in embryonic lethality. However, newborn mice die shortly after birth due to intracerebral hemorrhages, probably due to a disruption of the mechanical association of the central nervous parenchyma with cerebral microvessels.143

The αIIb β3 Integrin

This integrin is exclusively found expressed on the surface of megakaryocytes and platelets. It is essential for platelet aggregation and subsequent clot retraction. Therefore, this integrin provides firm adhesion and seems not to be involved in cell migration. However, during clot retraction, αIIbβ3 integrin dependent fibrin interactions is undergoing remodeling, reflecting in many ways the rearrangements of focal contacts at the rear of migrating cells (see below). Therefore, many aspects of integrin physiology first established for this integrin, such as activation induced unfolding, cytoplasmic adaptor binding and intracellular signaling, form the basis on which the behavior and function of other integrins can be predicted.144-150

Modulation of Integrin Affinity, Valency and Dynamics during Cell Migration

In order to understand the role of integrins during cell migration and to identify critical targets for therapeutical interventions, the behavior of integrin receptors during the formation, remodeling and dispersal of focal adhesions has to be determined and quantitatively analyzed. An important concept of integrin dependent adhesion encompasses the notion that integrin dependent adhesion is controlled by modulations of the affinity and the avidity of integrin receptors. Because the use of the term “avidity” has not been consistent,108 it is important to define the different integrin specific parameters that are involved in the modulation of size and mechanical strength of focal adhesion sites. Three different integrin dependent mechanisms can be distinguished that involve (i) integrin affinity modulations, (ii) changes in the valency of integrin dependent adhesions and (iii) modifications of the dynamic exchange rates of integrins within focal adhesion sites.

Integrin affinity modulations reflect the allosteric switch from the inactive integrin heterodimer to the fully unfolded extracellular ligand-binding receptor. Depending on the mode of integrin activation (inside-out versus outside-in), several differentially folded intermediates may exist in a dynamic equilibrium within the plasma membrane.145

Integrin valency modulations reflect the ability of integrin receptors to undergo lateral clustering within the plasma membrane in order to increase the number of individual integrin interactions and augment the overall cellular avidity towards extracellular ligands. Thus, the valency of integrin dependent binding is the result of “integrin packing density” multiplied by the “size” of focal adhesions and correlates with the overall strength and mechanical resistance of the link between the cell and the extracellular matrix.

Integrin dynamics reflect modulations of the temporal stability of integrins within focal adhesions. It is characterized by the time, expressed as a half-live, that integrin heterodimers persist within a given focal adhesion. The degree of integrin dynamics determines the remodeling capacity of focal adhesions.

In order for a cell to migrate and to form new focal complexes at the cell front and to remodel and disperse focal contacts at the cell rear, integrin affinity, valency and dynamics have to be regulated appropriately (see Fig. 1, Fig. 2). (i) At the cell front, integrins have to be in a high affinity state in order to bind to extracellular ligands encountered by filopodia and lamellipodia (high affinity). Once ligand is bound or recognized, rapid lateral clustering of integrins (increase in valency) is required to stabilize individual integrin contacts. In addition, a slow rate of integrin exchange (low dynamics) in newly formed focal complexes is essential to stabilize the actin cytoskeletal scaffold of filopodia and lamellipodia. (ii) Focal contacts that are localized at the level of the main cell body rapidly modulate their size and strength while maintaining a high degree of plasticity. Thus an elevated level of high-affinity integrins is required to rapidly respond to signals that modify the valency (size and density) of focal contacts. In addition, an increase in integrin dynamics provides focal contacts with the capacity to respond to mechanical changes in the cellular environment. (iii) At the cell rear, sliding and dispersal of focal contacts require an elevated level of integrin dynamics. The overall tendency of focal contacts to disperse at the rear of migrating cells, may suggest that the concentration of high-affinity integrins is reduced. In contrast, the polarized integrin assembly at the inner edge of sliding focal contacts requires a highly controlled regulation of integrin valency.

Figure 1. Signal dependent regulation of integrin affinity and valency.

Figure 1

Signal dependent regulation of integrin affinity and valency. Schematic representation of the equilibrium between low and high-affinity integrins and its regulation by the binding of extracellular ligands or intracellular adaptors (left hand side). High (more...)

Figure 2. Modulation of integrin affinity, valency and dynamics between the front and the rear of migrating cells.

Figure 2

Modulation of integrin affinity, valency and dynamics between the front and the rear of migrating cells. A) Schematic representation of integrin behavior in focal complexes versus focal contacts in migrating cells. Analysis of integrin dynamics in focal (more...)

Integrin Affinity Modulations during Cell Migration

Affinity Modulations at the Cell Front

Cell migration through a complex tissue environment intuitively requires the need for receptors that can sense and explore the newly encountered extracellular matrix. In contrast, rear detachment of migrating cells suggests that integrins have lost their affinity for substrate. Surprisingly, despite the availability of antibody reagents that specifically recognize high affinity forms of integrins, the detection of increased concentration of high affinity integrins at the front of migrating cells has been difficult. Symptomatic for this, only one report has so far demonstrated the accumulation of high affinity αvβ3 integrins at the front of migrating cell.151 Interestingly, the activation of the small GTPase Rac1, which is involved in the formation of lamellipodia is required for this polarized accumulation of high-affinity integrins.151 α4β1 integrins are also found at the leading edge of migrating cells, however, it is not known whether this integrin is retained their in a high affinity state.83,84 Similarly, it can only be hypothesized that the β1-integrins that colocalize with talin at the leading edge of lamellipodia in fish keratinocytes are in their high affinity configuration.152 In migrating leukocytes, RAPL that has been demonstrated to activate αLβ2 integrins, is localized and recruited to the front of migrating cells.104 Hence Rap1 dependent activation of RAPL in parallel with Rac1 signaling are involved in the polarized expression of high affinity integrins to the front of migrating cells.

Affinity Modulations at the Cell Rear

In dynamic focal contacts that are localized in the main cell body or towards the rear of the cell, the continuous incorporation and exchange of integrins suggests the presence of a local source of high affinity integrins. So far, it is not known whether integrins are laterally recruited from the surrounding plasma membrane or whether the newly incorporated high-affinity integrins are delivered to focal adhesions by vesicular transport.153 However, based on several independent experimental findings, it is possible that focal adhesions are able to create their own local source of high affinity integrins. This idea is essentially based on the crucial role of talin in the induction of the high affinity state of integrins.154,155 Integrins are activated by the binding of the head domain of talin to the β-integrin cytoplasmic tail.156 Importantly, this process is regulated by intracellular signaling, involving calpain-mediated proteolysis of talin.157 At the cell rear, increased rates of cell detachment are induced by MEKK signaling, resulting in calpain dependent proteolysis of focal adhesion proteins.158 Consistently, the expression of a calpain resistant form of talin renders focal contacts more static, suggesting that calpain mediated talin proteolysis is responsible for integrin dynamics rather than integrin activation.159 An alternative pathway of talin-dependent integrin activation involves phosphatidylinositol 4,5 bi-phosphate (PI(4,5)P2) binding to the talin-head domain. It has been demonstrated that PI(4,5)P2 binds to the head domain of talin which results in a change of the conformation of talin, resulting in integrin cytoplasmic tail binding and integrin activation.160,161 Talin is also able to recruit the PI(4,5)P2 synthesizing enzyme PIPKI γo focal adhesions.162,163 The recruitment of this kinase is critically required for focal adhesion formation. Thus, the local synthesis of PI(4,5)P2 in focal contacts may be instrumental in talin dependent integrin activation as well as the PI(4,5)P2 dependent formation of actin stress fibers.164

Valency Regulation of Integrins during Cell Migration

Valency Modulations in the Nano- and Micrometer Range

The modulation of the lateral association of integrin molecules during cell migration is crucial for the assembly of new focal adhesions at the cell front and their dispersal at the cell rear. Integrin clustering or an increase in valency, has been observed at two different scales. First, integrins form nanoclusters, consisting of a few integrin molecules that can only be demonstrated with specific methods relaying on resonance energy transfer such as FRET and BRET, or statistical methods such as image correlation microsocopy.97,144 It has been demonstrated that nano-clustering of integrins can be induced by multivalent ligands such as the bi-valent fibrinogen.144 Integrin nano-clusters may be critical for integrin dependent signaling and mechanical coupling of extracellular matrix to the actin cytoskeleton.165 It has been proposed that the critical limits, required for a mechanical coupling of extracellular ligands to the actin cytoskeleton is the clustering of three integrins. This threshold has been determined by a recombinant tri-meric fibronectin molecule.166

A second class of valency regulation occurs at the micrometer level. In fact, the microscopic observation of focal complexes reveals adhesion surfaces that do not exceed 1 μm in diameter. In contrast, streak like focal contacts in stationary or migrating cells can easily extend to 5-10 μm in length. Interestingly, while the size of focal complexes is rather uniform, the size of focal contacts is dependent on the degree of intracellular tension.34,167

Valency Regulation by Modulation of Integrin Density

In addition to a difference in their size, focal complexes and focal contacts can be distinguished by a low versus a high integrin packing density, respectively.34,97 In focal contacts, an acto-myosin dependent contraction of the actin-scaffold increases integrin density.27,34 This observation also confirmed the notion that the formation of focal complexes does not require myosin based actin cytoskeleton contraction and can form in a tension free environment in advancing filopodia or lamellipodia.33,34 Based on the different integrin packing densities between focal complexes and focal contacts, we have proposed that a limitation of the coating density of extracellular ligands would affect focal contact stability.27 Accordingly, the critical threshold of monomeric ligand density required for focal contact formation was initially determined to be between 120 and 440 nm.168 Recently, using an elegant method of ligand coating, this threshold was determined to be between 58 and 73 nm.169 Thus, cells can interact with, and migrate on substrates that allow the spacing between two integrins to be 73 nm or larger, however these cells will only be able to form focal contacts when integrin spacing is smaller or equivalent to 58 nm.169

Therefore, the acto-myosin driven increase in integrin density requires a reduction in the spacing of extracellular ligands. Accordingly, an increase in the tension of the extracellular matrix, such as in a stretched collagen gel, would enlarge the spacing of integrin ligands. This would directly reduce integrin valency in focal contacts, potentially inducing intracellular signaling pathways. It has been demonstrated that a 15% increase in the surface of a flexible substrate can induce FAK and paxillin recruitment to focal contacts.170 In turn, FAK activity is involved in the mechano-sensing of migrating fibroblasts.171 If fibroblasts are exposed to cyclic mechanical stress, a change in gene expression is induced.172 Interestingly, in this situation, Rho-kinase activity is critically required to transduce the externally mediated structural modulations of the actin cytoskeleton and focal contacts into altered gene expression.173 This suggests that the link between individual integrins and the actin cytoskeleton is critically involved in valency mediated signaling or perception of the elasticity of the extracellular environment. Accordingly, cells respond to a gradient of extracellular elasticity by directed migration towards the stiffer substrate, a behavior that has been named “durotaxis”.174,175

Hence, we can conclude that integrin valency modulations imposed by a nano or micro-patterned extracellular matrix are critically involved in cell spreading,169,176 the capacity to respond to extracellular mechanical perturbations165 and the sensing of the microelasticity of the extracellular environment.171,174,175

Dynamic Integrin Regulations during Cell Migration

Slow Integrin Dynamics at the Cell Front and Fast Dynamics at the Cell Rear

The first experiments that used dynamic methods to analyze the diffusion of integrin receptors in the plasma membrane were based on fluorescently labeled anti-integrin antibodies.177 These noninterfering antibodies were added to living cells and the diffusion and clustering of the antibody/integrin complexes was measured by fluorescence recovery after photobleaching (FRAP).178,179 In the combination with GFP labeled proteins, FRAP turned out to be an extremely powerful technique in order to analyze the association and dissociation dynamics of a complex macromolecular structure such as focal contacts. So far, only a few cytoplasmic adaptor proteins have been analyzed by this technique, nevertheless demonstrating the potential of this method to analyze the dynamic modulations of focal adhesions (e.g., ref. 180). In respect to integrins, we have used this technique to analyze integrin dynamics in the different types of focal adhesions that are formed in migrating cells.27,34 It became apparent that the organization of the actin cytoskeleton plays a crucial role in the integrin exchange dynamics of focal adhesions. In low-density integrin adhesion sites, representing focal complexes, αvβ3 integrin exchange rates are slow.34 In contrast, in focal contacts connected to actin stress fibers, αvβ3 integrin exchange is rapid, leading to a complete turnover of integrins within 10 minutes. 34 These observations have been made in focal contacts of highly mobile mouse melanoma cells. In contrast, the dynamics of αvβ3 integrin in focal contacts formed in stationary and highly contractile rat embryo fibroblasts is much slower.181 This dynamics is even slower, when these cells begin to express the α-smooth muscle actin isoform,181 assuming the contractile phenotype of myofibroblasts. In endothelial cells, another FRAP analysis of β3-GFP-integrin revealed that αvβ3 integrins trade places within a given focal contact. Interestingly, the intra-contact exchange rate is faster compared to the de-novo recruitment of integrins to focal contacts.182 This suggests that within dynamic focal contacts, individual integrins exchange their place continuously, reducing the need for integrin recruitment.

Based on these data, it is apparent that the different half-lives of integrins are only partially linked to the overall stability or persistence of a given focal adhesion. For example at the front of fast migrating melanoma cells, the turnover of focal complexes is faster than the respective exchange rates of the αvβ3 integrin. In this situation, the rapid turnover of focal complexes can be explained by the depolymerization of the actin scaffold towards the inner edge of the lamellipodium. In the main cell body however, focal contacts persist much longer than the measured half-live of the integrin exchange. This demonstrates that the modulation of integrin dynamics is a tool to change the “quality” of focal adhesions, making them firm or plastic. Although integrin dynamics may not directly influence the turnover of focal contacts, it is likely that the degree of integrin dynamics determines the capacity and rate of assembly and remodeling of focal complexes and or focal contacts.

Integrin Dynamics in Sliding Focal Contacts

We have previously proposed that the sliding of focal contacts is a macroscopic representation of the recruitment of integrins to the inner edge and their dispersal at the outer edge of sliding focal contacts. Therefore, the maximal speed of focal contact sliding is determined by the rate of integrin recruitment to the inner edge of a sliding contact. Unfortunately, the regulatory mechanisms that control the recruitment of integrin to the inner edge of sliding focal contacts are not well understood. However, it is likely that this issue is directly linked to the mechanisms that control integrin clustering.

Are Integrin Affinity and Valency Modulations Linked?

At the cell front, changes in integrin affinity and valency (integrin clustering), often occur simultaneously, hence it would be possible that integrin activation is directly linked to or actively induces integrin clustering. In fact, based on in vitro studies such a scenario was proposed for the αIIbβ3 integrin. Li and coworkers suggested that the activation induced unclasping of the integrin transmembrane domains183 allowed these isolated transmembrane domains to undergo homo-oligomerization with other α and β subunits. This transmembrane association would result in the formation of an inter-connected network of high affinity integrins, resulting in integrin clustering and increase in integrin valency. Thus, this result would predict that integrin affinity modulations for example induced by the binding of cytoplasmic adaptor proteins such as RAPL or talin (see above) would control the increase or decrease of integrin clustering in focal adhesions. Hence the control of the allosteric switch that regulates integrin activation would be of primordial importance for the behavior of individual focal adhesions.

Based on these data, we would predict that integrin activation by mutagenesis within the cytoplasmic or the extracellular domain would result in increased integrin clustering in living cells and modification of cell behavior. Accordingly the destruction of a salt bridge between the cytoplasmic domains of integrins (hinge region), results in integrin activation in CHO cells and increases tumor metastasis in carcinoma cells.130,148 A mutation in the extracellular ligand-binding region of β1-integrin increases integrin affinity and induces the formation of sarcomas, suggesting the importance for integrin affinity regulations in cell migration and differentiation.184 Similarly, the introduction of a “glycan wedge” that blocks the extracellular domain of αvβ3 integrin in the open configuration results in increased cell adhesion.185

Therefore, it is important to know whether affinity modulations of integrins have a direct or indirect impact on valency changes of integrins. Recently, the role of the integrin transmembrane domains in lateral integrin clustering has been analyzed by disulfide bond scanning of the exofacial side of the αIIbβ3 transmembrane domains. This analysis did not reveal a specific transmembrane mediated association of activated integrins in living cells.147 Furthermore, integrin activation by disrupting the respective association between the transmembrane domains of the α- and β-integrin did not increase integrin clustering.186 These observations are in apparent conflict with the visualized interaction between the stalk domains of purified integrins by electron-microscopy.183,187 In these integrin preparations, integrins form small clusters apparently linked by a region of the proteins that includes the transmembrane domain. However, since the detergent, used to purify integrins had to be removed prior to EM observation by rotary shadowing, it is not clear to which extent these integrin oligomers are due to the hydrophobic association of the transmembrane domains in a detergent free buffer. In fact, when purified αIIbβ3 integrins are re incorporated into vesicles or planar membranes, no apparent clustering of activated αIIbβ3 integrins can be observed when analyzed by electronmicroscopy of negatively stained vesicles.188 Nevertheless, within these reconstituted planar membranes, integrin clustering can be readily induced by the addition of the bivalent extracellular ligand fibrinogen.188 In conclusion, these data suggest that integrin activation is a prerequisite but by itself insufficient for integrin clustering. In order to induce integrin clustering, the interaction with multivalent extracellular ligands and/or intracellular adaptors is required.

Lateral Integrin Clustering and Adaptor Recruitment at the Front of Migrating Cells

Chronology of Integrin Clustering and Adaptor Recruitment

At the leading edge of migrating cells, activated integrins cluster to form focal complexes. Focal complexes consist of integrins and various adaptor proteins that provide the link between the integrin and the actin cytoskeleton. Recently the chronology of αvβ3 integrin clustering at the leading edge and respective adaptor protein recruitment was analyzed in migrating endothelial cells.85 This study revealed that clusters of αvβ3 integrins formed behind actin-rich regions in the lamellipodium. These integrin clusters, termed early focal complexes were positive for anti-phosphotyrosine antibodies, talin and paxillin but exhibited only low levels of vinculin and FAK.85 Older focal complexes additionally recruited FAK, vinculin, VASP, α-actinin and the Arp2/3 complex while zyxin was only localized to focal contacts.85 This temporal pattern of adaptor recruitment confirms older reports demonstrating the localization of talin and β1-integrins to the edge of protruding lamellipodia.152,189 This suggests that the Arp2/3 and vinculin dependent link of the actin cytoskeleton to focal complexes190 could be independently controlled from the clustering of integrins. Since talin, in addition to the integrin binding domains, exhibits also actin-binding sites, it could be the critical integrin adaptor that anchors extracellular ligand immobilized, clustered integrins to the actin cytoskeleton of the protruding lamellipodium.191

The Role of PI(4,5)P2 Lipids

However, there is still one missing piece in the puzzle of integrin clustering into focal complexes. Despite the importance of talin as a β3-integrin adaptor protein, talin is not sufficient to induce the clustering of activated integrins by itself. In fact, the sequestration of PI(4,5)P2 by the overexpression of the PH domain of PLCΔ161 inhibits talin recruitment to focal adhesions. Hence talin may require the PI(4,5)P2 lipid as a cofactor to bind to activated integrins and to link them to PI(4,5)P2 containing lipid domains. Moreover, the activation of vinculin and the cross-linking of F-actin to the plasma membrane by ERM proteins requires PI(4,5)P2.164 Furthermore, the phosphatidylinositol 4-phosphate 5-kinase α is a downstream effector of the small GTPase ARF6.192 The parallel activation of Rac1 and ARF6 are required for the formation of lamellipodia.193,194 Thus the combination of PI(4,5)P2 synthesis and actin polymerization at the leading edge of migrating cells create the subcellular environment that allows talin activation, integrin clustering and membrane associated actin polymerization, resulting in the formation of focal complexes.

Regulation of Integrin Clustering at the Cell Rear

In contrast to the cell front, which is under the influence of Rac1 and ARF6 signaling, RhoA signaling controls the formation of actin stress fibers and the formation of focal contacts. As mentioned above the integrin exchange rate in focal contacts is faster than in focal complexes. Because integrin dynamics are not influenced by integrin activation, it is likely that the rate of integrin turnover is regulated at the level of the linkage between the cytoplasmic tail of integrins and the actin cytoskeleton. Several intracellular signaling pathways have been associated with an increase in focal adhesion dynamics and increased migration rate. Integrin dependent FAK activation, c-src signaling and the MAPK pathway have all been implicated in the regulation of focal adhesion turnover.158,195-197 One of the crucial elements of this remodeling pathway is the activation of cytoplasmic protease μ-calpain.158,198-200 The calpain induced proteolytic cleavage of talin into an integrin binding head and actin binding tail domain may result in the destabilization of focal contacts. Interestingly, the expression of calpain insensitive talin stabilizes the cycle of focal contact assembly and disassembly.159 It will be interesting to determine, whether integrin dynamics are also slowed down in focal contacts containing protease resistant talin.

In conclusion, several intracellular signaling pathways converge at the cell front to firmly link activated integrins to the actin cytoskeleton, a process which requires adaptor proteins and PI(4,5)P2. At the cell rear, the continuous remodeling of the link between integrins and the actin cytoskeleton, allows a precise control over cell migration and regulated rear detachment.

Intracellular Integrin Transport

So far, the discussion of the role of integrins in cell migration has been limited to changes in integrin affinity, clustering and dynamics. Generally these mechanisms are differentially regulated between the front and the rear of migrating cells. In order for cell migration to persist, the maintenance of the anterior to posterior polarity is crucial. One important concept of cell polarity is the directed transport of proteins and membranes to the front of the cell. It has been proposed that cell motility is regulated by the transport of membranes and adhesion receptors from the rear to the front of migrating cells.201 As discussed above, some integrin receptors have been observed to get internalized at the cell rear and to be transported to the cell front.96

Detailed data have been gathered on the recycling of αvβ3 integrins in growth factor stimulated cells. It has been demonstrated that the recycling pathway of integrins is accelerated in PDGF treated cells. Instead of the recruitment of integrins from Rab11 positive late endosomes in nonstimulated cells, PDGF treatment induces the recycling of αvβ3 integrins from Rab4 positive early endosomes.153 Furthermore, it has been demonstrated that the PDK1 kinase that associates with the cytoplasmic tail of β3-integrins is required for this rapid integrin recycling, enabling cell spreading and migration.202 In addition, growth factor induced integrin recycling to the cell surface, requires the phosphorylation and inhibition of glycogen synthase kinase 3 (GSK-3) by protein kinase B/Akt.203 These data demonstrate the important link between growth factor induced formation of lamellipodia and the accelerated recycling of αvβ3 integrin to the front of migrating cells.

During PDGF stimulated integrin recycling, integrins reappear at the dorsal surface and accumulate only later at the cell front.153 It is therefore not clear whether integrins reach the front of migrating cells by vesicular transport, by lateral diffusion or by a different transport mechanism. The recent identification of the specific interaction of the cytoplasmic tail of β3 integrins with the FERM domain of myosin-X has suggests the existence of a powerful, myosin based integrin traffic. The over-expression of myosin-X induces the formation of filopodia in which integrins are colocalized with myosin-X to their tips.204 This suggests that αvβ3 integrins have been transported to the tips of filopodia by this motor protein. Since the integrin interacting FERM domain of myosin-X has homologies to the integrin binding FERM domain of talin, it is likely, that integrins can only be transported when their cytoplasmic tails are in an unclasped, active conformation. The subsequent release of αvβ3 integrins from myosin-X may be in response to a local competition with other integrin adaptor proteins such as talin, or may occur due to the mechanical interaction of the transported integrins with immobilized extracellular ligands. Myosin-X driven integrin delivery to the front of migrating cells may therefore occur along actin filaments bundles within filopodia. Interestingly, focal complexes in migrating endothelial cells form predominantly at the base of actin rich ridges,85 which could represent bona-fide myosin-X dependent integrin delivery pathways. Based on these data, specific integrin recycling and targeted delivery to the front of migrating cells are processes that are critically involved in the establishment of new integrin dependent contacts with the extracellular matrix.

Conclusion and Perspectives

We have learned that integrins play critical roles to control cell migration through extracellular matrix. Interestingly, different types of motile cells have acquired specific strategies to harvest the capacities of different types of integrins to undergo reversible ligand binding, in order to develop highly specific and tightly controlled mechanisms to direct cell migration through a complex tissue environment. It will remain a challenge to dissect the multitude of signaling pathways and to identify the hierarchies of protein-protein or protein-lipid interactions that control integrin clustering, dispersal and transport, essential steps for cell migration to occur.

Dedication

In memory of Martin Pfaff.

Acknowledgements

I would like to thank Drs Beat Imhof, Frederic Saltel and Michel Aurrand-Lions for discussions and sharing ideas. A special thanks to Monique Wehrle-Haller for helping me with the manuscript and the Swiss National Science Foundation for supporting my research (31-64000.00 and 3100A0-103805).

References

1.
Hynes RO. Integrins: Bidirectional, allosteric signaling machines. Cell. 2002;110(6):673–687. [PubMed: 12297042]
2.
Park KC, Rivero F, Meili R. et al. Rac regulation of chemotaxis and morphogenesis in Dictyostelium. EMBO J. 2004;23(-21):4177–4189. [PMC free article: PMC524383] [PubMed: 15470506]
3.
Funamoto S, Meili R, Lee S. et al. Spatial and temporal regulation of 3-phosphoinositides by PI 3-kinase and PTEN mediates chemotaxis. Cell. 2002;109(5):611–623. [PubMed: 12062104]
4.
Bretschneider T, Jonkman J, Kohler J. et al. Dynamic organization of the actin system in the motile cells of Dictyostelium. J Muscle Res Cell Motil. 2002;23(7-8):639–649. [PubMed: 12952063]
5.
Kreitmeier M, Gerisch G, Heizer C. et al. A talin homologue of Dictyostelium rapidly assembles at the leading edge of cells in response to chemoattractant. J Cell Biol. 1995;129(1):179–188. [PMC free article: PMC2120370] [PubMed: 7698984]
6.
Niewohner J, Weber I, Maniak M. et al. Talin-null cells of Dictyostelium are strongly defective in adhesion to particle and substrate surfaces and slightly impaired in cytokinesis. J Cell Biol. 1997;138(2):349–361. [PMC free article: PMC2138202] [PubMed: 9230077]
7.
Reber-Muller S, Studer R, Muller P. et al. Integrin and talin in the jellyfish Podocoryne carnea. Cell Biol Int. 2001;25(8):753–769. [PubMed: 11482899]
8.
Hughes AL. Evolution of the integrin alpha and beta protein families. J Mol Evol. 2001;52(1):63–72. [PubMed: 11139295]
9.
Brower DL, Brower SM, Hayward DC. et al. Molecular evolution of integrins: Genes encoding integrin beta subunits from a coral and a sponge. Proc Natl Acad Sci USA. 1997;94(17):9182–9187. [PMC free article: PMC23098] [PubMed: 9256456]
10.
Pancer Z, Kruse M, Muller I. et al. On the origin of Metazoan adhesion receptors: Cloning of integrin alpha subunit from the sponge Geodia cydonium. Mol Biol Evol. 1997;14(4):391–398. [PubMed: 9100369]
11.
Whittaker CA, Hynes RO. Distribution and evolution of von Willebrand/integrin A domains: Widely dispersed domains with roles in cell adhesion and elsewhere. Mol Biol Cell. 2002;13(10):3369–3387. [PMC free article: PMC129952] [PubMed: 12388743]
12.
Brown NH. Cell-cell adhesion via the ECM: Integrin genetics in fly and worm. Matrix Biol. 2000;19(3):191–201. [PubMed: 10936444]
13.
Bokel C, Brown NH. Integrins in development: Moving on, responding to, and sticking to the extracellular matrix. Dev Cell. 2002;3(3):311–321. [PubMed: 12361595]
14.
Ribeiro C, Petit V, Affolter M. Signaling systems, guided cell migration, and organogenesis: Insights from genetic studies in Drosophila. Dev Biol. 2003;260(1):1–8. [PubMed: 12885551]
15.
Niewiadomska P, Godt D, Tepass U. DE-Cadherin is required for intercellular motility during Drosophila oogenesis. J Cell Biol. 1999;144(3):533–547. [PMC free article: PMC2132905] [PubMed: 9971747]
16.
Devenport D, Brown NH. Morphogenesis in the absence of integrins: Mutation of both Drosophila beta subunits prevents midgut migration. Development. 2004;131(21):5405–5415. [PubMed: 15469969]
17.
Rogalski TM, Mullen GP, Gilbert MM. et al. The UNC-112 gene in Caenorhabditis elegans encodes a novel component of cell-matrix adhesion structures required for integrin localization in the muscle cell membrane. J Cell Biol. 2000;150(1):253–264. [PMC free article: PMC2185566] [PubMed: 10893272]
18.
Brown NH, Gregory SL, Rickoll WL. et al. Talin is essential for integrin function in Drosophila. Dev Cell. 2002;3(4):569–579. [PubMed: 12408808]
19.
Cram EJ, Clark SG, Schwarzbauer JE. Talin loss-of-function uncovers roles in cell contractility and migration in C. jelegans. J Cell Sci. 2003;116(Pt 19):3871–3878. [PubMed: 12915588]
20.
Starz-Gaiano M, Montell DJ. Genes that drive invasion and migration in Drosophila. Curr Opin Genet Dev. 2004;14(1):86–91. [PubMed: 15108810]
21.
Somogyi K, Rorth P. Evidence for tension-based regulation of Drosophila MAL and SRF during invasive cell migration. Dev Cell. 2004;7(1):85–93. [PubMed: 15239956]
22.
Pinheiro EM, Montell DJ. Requirement for Par-6 and Bazooka in Drosophila border cell migration. Development. 2004;131(21):5243–5251. [PubMed: 15456726]
23.
Lee M, Cram EJ, Shen B. et al. Roles for beta(pat-3) integrins in development and function of Caenorhabditis elegans muscles and gonads. J Biol Chem. 2001;276(39):36404–36410. [PubMed: 11473126]
24.
Weston JA, Abercrombie M. Cell mobility in fused homo- and heteronomic tissue fragments. J Exp Zool. 1967;164(3):317–323. [PubMed: 6035668]
25.
Abercrombie M, Dunn GA, Heath JP. The shape and movement of fibroblasts in culture. Soc Gen Physiol Ser. 1977;32:57–70. [PubMed: 333596]
26.
Wehrle-Haller B, Imhof BA. Actin, microtubules and focal adhesion dynamics during cell migration. Int J Biochem and Cell Biol. 2003;35:39–50. [PubMed: 12467646]
27.
Wehrle-Haller B, Imhof BA. The inner lives of focal adhesions. Trends Cell Biol. 2002;12(8):382–389. [PubMed: 12191915]
28.
Friedl P, Borgmann S, Brocker EB. Amoeboid leukocyte crawling through extracellular matrix: Lessons from the Dictyostelium paradigm of cell movement. J Leukoc Biol. 2001;70(4):491–509. [PubMed: 11590185]
29.
Cukierman E, Pankov R, Yamada KM. Cell interactions with three-dimensional matrices. Curr Opin Cell Biol. 2002;14(5):633–639. [PubMed: 12231360]
30.
Small JV, Stradal T, Vignal E. et al. The lamellipodium: Where motility begins. Trends Cell Biol. 2002;12(3):112–120. [PubMed: 11859023]
31.
Borisy GG, Svitkina TM. Actin machinery: Pushing the envelope. Curr Opin Cell Biol. 2000;12(1):104–112. [PubMed: 10679366]
32.
Geiger B, Bershadsky A. Assembly and mechanosensory function of focal contacts. Curr Opin Cell Biol. 2001;13(5):584–592. [PubMed: 11544027]
33.
Riveline D, Zamir E, Balaban NQ. et al. Focal contacts as mechanosensors: Externally applied local mechanical force induces growth of focal contacts by an mDia1-dependent and ROCK- independent mechanism. J Cell Biol. 2001;153(6):1175–1186. [PMC free article: PMC2192034] [PubMed: 11402062]
34.
Ballestrem C, Hinz B, Imhof BA. et al. Marching at the front and dragging behind: Differential alphaVbeta3- Integrin turnover regulates focal adhesion behavior. J Cell Biol. 2001;155(7):1319–1332. [PMC free article: PMC2199321] [PubMed: 11756480]
35.
Oliver T, Dembo M, Jacobson K. Separation of propulsive and adhesive traction stresses in locomoting keratocytes. J Cell Biol. 1999;145(3):589–604. [PMC free article: PMC2185069] [PubMed: 10225959]
36.
Lee J, Leonard M, Oliver T. et al. Traction forces generated by locomoting keratocytes. J Cell Biol. 1994;127(6 Pt 2):1957–1964. [PMC free article: PMC2120302] [PubMed: 7806573]
37.
Dembo M, Wang YL. Stresses at the cell-to-substrate interface during locomotion of fibroblasts. Biophys J. 1999;76(4):2307–2316. [PMC free article: PMC1300203] [PubMed: 10096925]
38.
Anderson KI, Cross R. Contact dynamics during keratocyte motility. Curr Biol. 2000;10(5):253–260. [PubMed: 10712904]
39.
Chen WT. Mechanism of retraction of the trailing edge during fibroblast movement. J Cell Biol. 1981;90(1):187–200. [PMC free article: PMC2111832] [PubMed: 7195906]
40.
Palecek SP, Huttenlocher A, Horwitz AF. et al. Physical and biochemical regulation of integrin release during rear detachment of migrating cells. J Cell Sci. 1998;111(Pt 7):929–940. [PubMed: 9490637]
41.
Palecek SP, Loftus JC, Ginsberg MH. et al. Integrin-ligand binding properties govern cell migration speed through cell-substratum adhesiveness. Nature. 1997;385(6616):537–540. [PubMed: 9020360]
42.
Smilenov LB, Mikhailov A, Pelham RJ. et al. Focal adhesion motility revealed in stationary fibroblasts [In Process Citation] Science. 1999;286(5442):1172–1174. [PubMed: 10550057]
43.
Hong YK, Lange-Asschenfeldt B, Velasco P. et al. VEGF-A promotes tissue repair-associated lymphatic vessel formation via VEGFR-2 and the alpha1beta1 and alpha2beta1 integrins. FASEB J. 2004;18(10):1111–1113. [PubMed: 15132990]
44.
Kagami S, Kondo S, Loster K. et al. Alpha1beta1 integrin-mediated collagen matrix remodeling by rat mesangial cells is differentially regulated by transforming growth factor-beta and platelet-derived growth factor-BB. J Am Soc Nephrol. 1999;10(4):779–789. [PubMed: 10203362]
45.
Ferrero E, Fabbri M, Poggi A. et al. Tumor-driven matrix invasion by infiltrating lymphocytes: Involvement of the alpha1 integrin I-domain. Eur J Immunol. 1998;28(8):2530–2536. [PubMed: 9710230]
46.
Stricker TP, Dumin JA, Dickeson SK. et al. Structural analysis of the alpha(2) integrin I domain/ procollagenase-1 (matrix metalloproteinase-1) interaction. J Biol Chem. 2001;276(31):29375–29381. [PubMed: 11359774]
47.
Camper L, Holmvall K, Wangnerud C. et al. Distribution of the collagen-binding integrin alpha10beta1 during mouse development. Cell Tissue Res. 2001;306(1):107–116. [PubMed: 11683172]
48.
Bengtsson T, Aszodi A, Nicolae C. et al. Loss of alpha10beta1 integrin expression leads to moderate dysfunction of growth plate chondrocytes. J Cell Sci. 2005;118(Pt 5):929–936. [PubMed: 15713743]
49.
Popova SN, Rodriguez-Sanchez B, Liden A. et al. The mesenchymal alpha11beta1 integrin attenuates PDGF-BB-stimulated chemotaxis of embryonic fibroblasts on collagens. Dev Biol. 2004;270(2):427–442. [PubMed: 15183724]
50.
Tiger CF, Fougerousse F, Grundstrom G. et al. alpha11beta1 integrin is a receptor for interstitial collagens involved in cell migration and collagen reorganization on mesenchymal nonmuscle cells. Dev Biol. 2001;237(1):116–129. [PubMed: 11518510]
51.
Schmid RS, Anton ES. Role of integrins in the development of the cerebral cortex. Cereb Cortex. 2003;13(3):219–224. [PubMed: 12571112]
52.
Schmid RS, Shelton S, Stanco A. et al. alpha3beta1 integrin modulates neuronal migration and placement during early stages of cerebral cortical development. Development. 2004;131(24):6023–6031. [PubMed: 15537685]
53.
Sanada K, Gupta A, Tsai LH. Disabled-1-regulated adhesion of migrating neurons to radial glial fiber contributes to neuronal positioning during early corticogenesis. Neuron. 2004;42(2):197–211. [PubMed: 15091337]
54.
Yoshimasu T, Sakurai T, Oura S. et al. Increased expression of integrin alpha3beta1 in highly brain metastatic subclone of a human nonsmall cell lung cancer cell line. Cancer Sci. 2004;95(2):142–148. [PubMed: 14965364]
55.
Tsuji T, Kawada Y, Kai-Murozono M. et al. Regulation of melanoma cell migration and invasion by laminin-5 and alpha3beta1 integrin (VLA-3). Clin Exp Metastasis. 2002;19(2):127–134. [PubMed: 11964076]
56.
Sordat I, Decraene C, Silvestre T. et al. Complementary DNA arrays identify CD63 tetraspanin and alpha3 integrin chain as differentially expressed in low and high metastatic human colon carcinoma cells. Lab Invest. 2002;82(12):1715–1724. [PubMed: 12480921]
57.
Pulkkinen L, Kimonis VE, Xu Y. et al. Homozygous alpha6 integrin mutation in junctional epidermolysis bullosa with congenital duodenal atresia. Hum Mol Genet. 1997;6(5):669–674. [PubMed: 9158140]
58.
Pulkkinen L, Rouan F, Bruckner-Tuderman L. et al. Novel ITGB4 mutations in lethal and nonlethal variants of epidermolysis bullosa with pyloric atresia: Missense versus nonsense. Am J Hum Genet. 1998;63(5):1376–1387. [PMC free article: PMC1377547] [PubMed: 9792864]
59.
Pulkkinen L, Uitto J. Mutation analysis and molecular genetics of epidermolysis bullosa. Matrix Biol. 1999;18(1):29–42. [PubMed: 10367729]
60.
Koster J, van WilpeS, Kuikman I. et al. Role of binding of plectin to the integrin beta4 subunit in the assembly of hemidesmosomes. Mol Biol Cell. 2004;15(3):1211–1223. [PMC free article: PMC363110] [PubMed: 14668477]
61.
Jonkman MF, Pas HH, Nijenhuis M. et al. Deletion of a cytoplasmic domain of integrin beta4 causes epidermolysis bullosa simplex. J Invest Dermatol. 2002;119(6):1275–1281. [PubMed: 12485428]
62.
Ashton GH, McLean WH, South AP. et al. Recurrent mutations in kindlin-1, a novel keratinocyte focal contact protein, in the autosomal recessive skin fragility and photosensitivity disorder, Kindler syndrome. J Invest Dermatol. 2004;122(1):78–83. [PubMed: 14962093]
63.
Siegel DH, Ashton GH, Penagos HG. et al. Loss of kindlin-1, a human homolog of the Caenorhabditis elegans actin-extracellular-matrix linker protein UNC-112, causes Kindler syndrome. Am J Hum Genet. 2003;73(1):174–187. [PMC free article: PMC1180579] [PubMed: 12789646]
64.
Kloeker S, Major MB, Calderwood DA. et al. The Kindler syndrome protein is regulated by transforming growth factor-beta and involved in integrin-mediated adhesion. J Biol Chem. 2004;279(8):6824–6833. [PubMed: 14634021]
65.
De ArcangelisA, Georges-Labouesse E. Integrin and ECM functions: Roles in vertebrate development. Trends Genet. 2000;16(9):389–395. [PubMed: 10973067]
66.
Georges-Labouesse E, Mark M, Messaddeq N. et al. Essential role of alpha 6 integrins in cortical and retinal lamination. Curr Biol. 1998;8(17):983–986. [PubMed: 9742403]
67.
Graus-Porta D, Blaess S, Senften M. et al. Beta1-class integrins regulate the development of laminae and folia in the cerebral and cerebellar cortex. Neuron. 2001;31(3):367–379. [PubMed: 11516395]
68.
Dangerfield JP, Wang S, Nourshargh S. Blockade of alpha6 integrin inhibits IL-1beta- but not TNF-alpha-induced neutrophil transmigration in vivo. J Leukoc Biol. 2005;77(2):159–165. [PubMed: 15539457]
69.
Strachan LR, Condic ML. Cranial neural crest recycle surface integrins in a substratum-dependent manner to promote rapid motility. J Cell Biol. 2004;167(3):545–554. [PMC free article: PMC2172496] [PubMed: 15520227]
70.
Hayashi YK, Chou FL, Engvall E. et al. Mutations in the integrin alpha7 gene cause congenital myopathy. Nat Genet. 1998;19(1):94–97. [PubMed: 9590299]
71.
Mayer U, Saher G, Fassler R. et al. Absence of integrin alpha 7 causes a novel form of muscular dystrophy. Nat Genet. 1997;17(3):318–323. [PubMed: 9354797]
72.
Pegoraro E, Cepollaro F, Prandini P. et al. Integrin alpha 7 beta 1 in muscular dystrophy/myopathy of unknown etiology. Am J Pathol. 2002;160(6):2135–2143. [PMC free article: PMC1850814] [PubMed: 12057917]
73.
Kaariainen M, Kaariainen J, Jarvinen TL. et al. Integrin and dystrophin associated adhesion protein complexes during regeneration of shearing-type muscle injury. Neuromuscul Disord. 2000;10(2):121–132. [PubMed: 10714587]
74.
Kaariainen M, Nissinen L, Kaufman S. et al. Expression of alpha7beta1 integrin splicing variants during skeletal muscle regeneration. Am J Pathol. 2002;161(3):1023–1031. [PMC free article: PMC1867267] [PubMed: 12213731]
75.
Burkin DJ, Wallace GQ, Milner DJ. et al. Transgenic expression of alpha7beta1 integrin maintains muscle integrity, increases regenerative capacity, promotes hypertrophy, and reduces cardiomyopathy in dystrophic mice. Am J Pathol. 2005;166(1):253–263. [PMC free article: PMC1602287] [PubMed: 15632017]
76.
Burkin DJ, Wallace GQ, Nicol KJ. et al. Enhanced expression of the alpha 7 beta 1 integrin reduces muscular dystrophy and restores viability in dystrophic mice. J Cell Biol. 2001;152(6):1207–1218. [PMC free article: PMC2199213] [PubMed: 11257121]
77.
Arroyo AG, Yang JT, Rayburn H. et al. Alpha4 integrins regulate the proliferation/differentiation balance of multilineage hematopoietic progenitors in vivo. Immunity. 1999;11(5):555–566. [PubMed: 10591181]
78.
Taooka Y, Chen J, Yednock T. et al. The integrin alpha9beta1 mediates adhesion to activated endothelial cells and transendothelial neutrophil migration through interaction with vascular cell adhesion molecule-1. J Cell Biol. 1999;145(2):413–420. [PMC free article: PMC2133104] [PubMed: 10209034]
79.
Kil SH, Krull CE, Cann G. et al. The alpha4 subunit of integrin is important for neural crest cell migration. Dev Biol. 1998;202(1):29–42. [PubMed: 9758701]
80.
Testaz S, Duband JL. Central role of the alpha4beta1 integrin in the coordination of avian truncal neural crest cell adhesion, migration, and survival. Dev Dyn. 2001;222(2):127–140. [PubMed: 11668592]
81.
von AndrianUH, Engelhardt B. Alpha4 integrins as therapeutic targets in autoimmune disease. N Engl J Med. 2003;348(1):68–72. [PubMed: 12510047]
82.
Kassner PD, Alon R, Springer TA. et al. Specialized functional properties of the integrin alpha 4 cytoplasmic domain. Mol Biol Cell. 1995;6(6):661–674. [PMC free article: PMC301227] [PubMed: 7579686]
83.
Liu S, Thomas SM, Woodside DG. et al. Binding of paxillin to alpha4 integrins modifies integrin-dependent biological responses. Nature. 1999;402(6762):676–681. [PubMed: 10604475]
84.
Goldfinger LE, Han J, Kiosses WB. et al. Spatial restriction of alpha4 integrin phosphorylation regulates lamellipodial stability and alpha4beta1-dependent cell migration. J Cell Biol. 2003;162(4):731–741. [PMC free article: PMC2173787] [PubMed: 12913113]
85.
Zaidel-Bar R, Ballestrem C, Kam Z. et al. Early molecular events in the assembly of matrix adhesions at the leading edge of migrating cells. J Cell Sci. 2003;116(Pt 22):4605–4613. [PubMed: 14576354]
86.
Nishiya N, Kiosses WB, Han J. et al. An alpha(4) integrin-paxillin-Arf-GAP complex restricts Rac activation to the leading edge of migrating cells. Nat Cell Biol. 2005;7(4):343–352. [PubMed: 15793570]
87.
Rose DM, Liu S, Woodside DG. et al. Paxillin binding to the alpha 4 integrin subunit stimulates LFA-1 (integrin alpha L beta 2)-dependent T cell migration by augmenting the activation of focal adhesion kinase/proline-rich tyrosine kinase-2. J Immunol. 2003;170(12):5912–5918. [PubMed: 12794117]
88.
Young BA, Taooka Y, Liu S. et al. The cytoplasmic domain of the integrin alpha9 subunit requires the adaptor protein paxillin to inhibit cell spreading but promotes cell migration in a paxillin-independent manner. Mol Biol Cell. 2001;12(10):3214–3225. [PMC free article: PMC60168] [PubMed: 11598204]
89.
Chen C, Young BA, Coleman CS. et al. Spermidine/spermine N1-acetyltransferase specifically binds to the integrin alpha9 subunit cytoplasmic domain and enhances cell migration. J Cell Biol. 2004;167(1):161–170. [PMC free article: PMC2172529] [PubMed: 15479742]
90.
Huang XZ, Wu JF, Ferrando R. et al. Fatal bilateral chylothorax in mice lacking the integrin alpha9beta1. Mol Cell Biol. 2000;20(14):5208–5215. [PMC free article: PMC85969] [PubMed: 10866676]
91.
Vlahakis NE, Young BA, Atakilit A. et al. The lymphangiogenic vascular endothelial growth factors VEGF-C and -D are ligands for the integrin alpha9beta1. J Biol Chem. 2005;280(6):4544–4552. [PMC free article: PMC1368959] [PubMed: 15590642]
92.
Zamir E, Katz M, Posen Y. et al. Dynamics and segregation of cell-matrix adhesions in cultured fibroblasts. Nat Cell Biol. 2000;2(4):191–196. [PubMed: 10783236]
93.
Pankov R, Cukierman E, Katz BZ. et al. Integrin dynamics and matrix assembly: Tensin-dependent translocation of alpha(5)beta(1) integrins promotes early fibronectin fibrillogenesis. J Cell Biol. 2000;148(5):1075–1090. [PMC free article: PMC2174533] [PubMed: 10704455]
94.
Zajaczkowski MB, Cukierman E, Galbraith CG. et al. Cell-matrix adhesions on poly(vinyl alcohol) hydrogels. Tissue Eng. 2003;9(3):525–533. [PubMed: 12857420]
95.
Clark K, Pankov R, Travis MA. et al. A specific alpha5beta1-integrin conformation promotes directional integrin translocation and fibronectin matrix formation. J Cell Sci. 2005;118(Pt 2):291–300. [PMC free article: PMC3329624] [PubMed: 15615773]
96.
Laukaitis CM, Webb DJ, Donais K. et al. Differential dynamics of alpha 5 integrin, paxillin, and alpha-actinin during formation and disassembly of adhesions in migrating cells. J Cell Biol. 2001;153(7):1427–1440. [PMC free article: PMC2150721] [PubMed: 11425873]
97.
Wiseman PW, Brown CM, Webb DJ. et al. Spatial mapping of integrin interactions and dynamics during cell migration by image correlation microscopy. J Cell Sci. 2004;117(Pt 23):5521–5534. [PubMed: 15479718]
98.
Muller U, Wang D, Denda S. et al. Integrin alpha8beta1 is critically important for epithelial-mesenchymal interactions during kidney morphogenesis. Cell. 1997;88(5):603–613. [PMC free article: PMC2711892] [PubMed: 9054500]
99.
Brandenberger R, Schmidt A, Linton J. et al. Identification and characterization of a novel extracellular matrix protein nephronectin that is associated with integrin alpha8beta1 in the embryonic kidney. J Cell Biol. 2001;154(2):447–458. [PMC free article: PMC2150762] [PubMed: 11470831]
100.
Lu M, Munger JS, Steadele M. et al. Integrin alpha8beta1 mediates adhesion to LAP-TGFbeta1. J Cell Sci. 2002;115(Pt 23):4641–4648. [PubMed: 12415008]
101.
Bouzeghrane F, Mercure C, Reudelhuber TL. et al. Alpha8beta1 integrin is upregulated in myofibroblasts of fibrotic and scarring myocardium. J Mol Cell Cardiol. 2004;36(3):343–353. [PubMed: 15010273]
102.
Imhof BA, Aurrand-Lions M. Adhesion mechanisms regulating the migration of monocytes. Nat Rev Immunol. 2004;4(6):432–444. [PubMed: 15173832]
103.
Katagiri K, Ohnishi N, Kabashima K. et al. Crucial functions of the Rap1 effector molecule RAPL in lymphocyte and dendritic cell trafficking. Nat Immunol. 2004;5:1045–1051. [PubMed: 15361866]
104.
Katagiri K, Maeda A, Shimonaka M. et al. RAPL, a Rap1-binding molecule that mediates Rap1-induced adhesion through spatial regulation of LFA-1. Nat Immunol. 2003;4:741–748. [PubMed: 12845325]
105.
Bos JL, de RooijJ, Reedquist KA. Rap1 signalling: Adhering to new models. Nat Rev Mol Cell Biol. 2001;2(5):369–377. [PubMed: 11331911]
106.
de BruynKM, Rangarajan S, Reedquist KA. et al. The small GTPase Rap1 is required for Mn(2+)- and antibody-induced LFA-1- and VLA-4-mediated cell adhesion. J Biol Chem. 2002;277(33):29468–29476. [PubMed: 12171996]
107.
Tohyama Y, Katagiri K, Pardi R. et al. The critical cytoplasmic regions of the alphaL/beta2 integrin in Rap1-induced adhesion and migration. Mol Biol Cell. 2003;14(6):2570–2582. [PMC free article: PMC194904] [PubMed: 12808052]
108.
Carman CV, Springer TA. Integrin avidity regulation: Are changes in affinity and conformation underemphasized? Curr Opin Cell Biol. 2003;15(5):547–556. [PubMed: 14519389]
109.
Kim M, Carman CV, Springer TA. Bidirectional transmembrane signaling by cytoplasmic domain separation in integrins. Science. 2003;301(5640):1720–1725. [PubMed: 14500982]
110.
Niggli V. Rho-kinase in human neutrophils: A role in signalling for myosin light chain phosphorylation and cell migration. FEBS Lett. 1999;445(1):69–72. [PubMed: 10069376]
111.
Eddy RJ, Pierini LM, Matsumura F. et al. Ca2+-dependent myosin II activation is required for uropod retraction during neutrophil migration. J Cell Sci. 2000;113(Pt 7):1287–1298. [PubMed: 10704379]
112.
Lawson MA, Maxfield FR. Ca(2+)- and calcineurin-dependent recycling of an integrin to the front of migrating neutrophils. Nature. 1995;377(6544):75–79. [PubMed: 7544874]
113.
Pierini LM, Lawson MA, Eddy RJ. et al. Oriented endocytic recycling of alpha5beta1 in motile neutrophils. Blood. 2000;95(8):2471–2480. [PubMed: 10753823]
114.
Fabbri M, Fumagalli L, Bossi G. et al. A tyrosine-based sorting signal in the beta2 integrin cytoplasmic domain mediates its recycling to the plasma membrane and is required for ligand-supported migration. EMBO J. 1999;18(18):4915–4925. [PMC free article: PMC1171563] [PubMed: 10487744]
115.
Wagner N, Lohler J, Kunkel EJ. et al. Critical role for beta7 integrins in formation of the gut-associated lymphoid tissue. Nature. 1996;382(6589):366–370. [PubMed: 8684468]
116.
Schon MP, Schon M, Warren HB. et al. Cutaneous inflammatory disorder in integrin alphaE (CD103)-deficient mice. J Immunol. 2000;165(11):6583–6589. [PubMed: 11086102]
117.
Kellersmann R, Lazarovits A, Grant D. et al. Monoclonal antibody against beta7 integrins, but not beta7 deficiency, attenuates intestinal allograft rejection in mice. Transplantation. 2002;74(9):1327–1334. [PubMed: 12451274]
118.
Sun X, Qiao H, Shi J. et al. Beta7 integrins contribute to skin graft rejection. Transplantation. 2002;74(8):1202–1203. [PubMed: 12438975]
119.
Davis GE. The Mac-1 and p150,95 beta 2 integrins bind denatured proteins to mediate leukocyte cell-substrate adhesion. Exp Cell Res. 1992;200(2):242–252. [PubMed: 1572393]
120.
Vorup-Jensen T, Carman CV, Shimaoka M. et al. Exposure of acidic residues as a danger signal for recognition of fibrinogen and other macromolecules by integrin alphaXbeta2. Proc Natl Acad Sci USA. 2005;102(5):1614–1619. [PMC free article: PMC547869] [PubMed: 15665082]
121.
Kindzelskii AL, Laska ZO, Todd IIIrd RF. et al. Urokinase-type plasminogen activator receptor reversibly dissociates from complement receptor type 3 (alpha M beta 2' CD11b/CD18) during neutrophil polarization. J Immunol. 1996;156(1):297–309. [PubMed: 8598477]
122.
Kindzelskii AL, Eszes MM, Todd IIIrd RF. et al. Proximity oscillations of complement type 4 (alphaX beta2) and urokinase receptors on migrating neutrophils. Biophys J. 1997;73(4):1777–1784. [PMC free article: PMC1181078] [PubMed: 9336173]
123.
Mabon PJ, Weaver LC, Dekaban GA. Inhibition of monocyte/macrophage migration to a spinal cord injury site by an antibody to the integrin alphaD: A potential new anti-inflammatory treatment. Exp Neurol. 2000;166(1):52–64. [PubMed: 11031083]
124.
Law DA, DeGuzman FR, Heiser P. et al. Integrin cytoplasmic tyrosine motif is required for outside-in alphaIIbbeta3 signalling and platelet function. Nature. 1999;401(6755):808–811. [PubMed: 10548108]
125.
Boettiger D, Huber F, Lynch L. et al. Activation of alpha(v)beta3-vitronectin binding is a multistage process in which increases in bond strength are dependent on Y747 and Y759 in the cytoplasmic domain of beta3. Mol Biol Cell. 2001;12(5):1227–1237. [PMC free article: PMC34580] [PubMed: 11359918]
126.
Brooks PC, Clark RA, Cheresh DA. Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science. 1994;264(5158):569–571. [PubMed: 7512751]
127.
Eliceiri BP, Cheresh DA. Role of alpha v integrins during angiogenesis. Cancer J. 2000;6(Suppl 3):S245–249. [PubMed: 10874494]
128.
Filardo EJ, Brooks PC, Deming SL. et al. Requirement of the NPXY motif in the integrin beta 3 subunit cytoplasmic tail for melanoma cell migration in vitro and in vivo. J Cell Biol. 1995;130(2):441–450. [PMC free article: PMC2199943] [PubMed: 7542248]
129.
Bao W, Stromblad S. Integrin alphav-mediated inactivation of p53 controls a MEK1-dependent melanoma cell survival pathway in three-dimensional collagen. J Cell Biol. 2004;167(4):745–756. [PMC free article: PMC2172581] [PubMed: 15557124]
130.
Felding-Habermann B, O'Toole TE, Smith JW. et al. Integrin activation controls metastasis in human breast cancer. Proc Natl Acad Sci USA. 2001;98(4):1853–1858. [PMC free article: PMC29346] [PubMed: 11172040]
131.
Brooks PC, Montgomery AM, Rosenfeld M. et al. Integrin alpha v beta 3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell. 1994;79(7):1157–1164. [PubMed: 7528107]
132.
Gutheil JC, Campbell TN, Pierce PR. et al. Targeted antiangiogenic therapy for cancer using Vitaxin: A humanized monoclonal antibody to the integrin alphavbeta3. Clin Cancer Res. 2000;6(8):3056–3061. [PubMed: 10955784]
133.
Buckley CD, Pilling D, Henriquez NV. et al. RGD peptides induce apoptosis by direct caspase-3 activation. Nature. 1999;397(6719):534–539. [PubMed: 10028971]
134.
Reynolds LE, Wyder L, Lively JC. et al. Enhanced pathological angiogenesis in mice lacking beta3 integrin or beta3 and beta5 integrins. Nat Med. 2002;8(1):27–34. [PubMed: 11786903]
135.
Lane NE, Yao W, Nakamura MC. et al. Mice lacking the integrin beta5 subunit have accelerated osteoclast maturation and increased activity in the estrogen-deficient state. J Bone Miner Res. 2005;20(1):58–66. [PubMed: 15619670]
136.
Huang X, Griffiths M, Wu J. et al. Normal development, wound healing, and adenovirus susceptibility in beta5-deficient mice. Mol Cell Biol. 2000;20(3):755–759. [PMC free article: PMC85191] [PubMed: 10629031]
137.
Filardo EJ, Deming SL, Cheresh DA. Regulation of cell migration by the integrin beta subunit ectodomain. J Cell Sci. 1996;109(Pt 6):1615–1622. [PubMed: 8799848]
138.
Memmo LM, McKeown-Longo P. The alphavbeta5 integrin functions as an endocytic receptor for vitronectin. J Cell Sci. 1998;111(Pt 4):425–433. [PubMed: 9443892]
139.
Nandrot EF, Kim Y, Brodie SE. et al. Loss of synchronized retinal phagocytosis and age-related blindness in mice lacking alphavbeta5 integrin. J Exp Med. 2004;200(12):1539–1545. [PMC free article: PMC2211990] [PubMed: 15596525]
140.
Huang X, Wu J, Spong S. et al. The integrin alphavbeta6 is critical for keratinocyte migration on both its known ligand, fibronectin, and on vitronectin. J Cell Sci. 1998;111(Pt 15):2189–2195. [PubMed: 9664040]
141.
Huang X, Wu J, Zhu W. et al. Expression of the human integrin beta6 subunit in alveolar type II cells and bronchiolar epithelial cells reverses lung inflammation in beta6 knockout mice. Am J Respir Cell Mol Biol. 1998;19(4):636–642. [PubMed: 9761761]
142.
Nishimura SL, Boylen KP, Einheber S. et al. Synaptic and glial localization of the integrin alphavbeta8 in mouse and rat brain. Brain Res. 1998;791(1-2):271–282. [PubMed: 9593935]
143.
McCarty JH, Monahan-Earley RA, Brown LF. et al. Defective associations between blood vessels and brain parenchyma lead to cerebral hemorrhage in mice lacking alphav integrins. Mol Cell Biol. 2002;22(21):7667–7677. [PMC free article: PMC135679] [PubMed: 12370313]
144.
Buensuceso C, de VirgilioM, Shattil SJ. Detection of integrin alpha IIbbeta 3 clustering in living cells. J Biol Chem. 2003;278(17):15217–15224. [PubMed: 12595537]
145.
Xiao T, Takagi J, Coller BS. et al. Structural basis for allostery in integrins and binding to fibrinogen-mimetic therapeutics. Nature. 2004;432:59–67. [PMC free article: PMC4372090] [PubMed: 15378069]
146.
Vinogradova O, Velyvis A, Velyviene A. et al. A structural mechanism of integrin alpha(IIb)beta(3) "inside-out" activation as regulated by its cytoplasmic face. Cell. 2002;110(5):587–597. [PubMed: 12230976]
147.
Luo BH, Springer TA, Takagi J. A specific interface between integrin transmembrane helices and affinity for ligand. PLoS Biol. 2004;2:e153. [PMC free article: PMC423134] [PubMed: 15208712]
148.
Hughes PE, Diaz-Gonzalez F, Leong L. et al. Breaking the integrin hinge. A defined structural constraint regulates integrin signaling. J Biol Chem. 1996;271(12):6571–6574. [PubMed: 8636068]
149.
Bertoni A, Tadokoro S, Eto K. et al. Relationships between Rap1b, affinity modulation of integrin alpha IIbbeta 3, and the actin cytoskeleton. J Biol Chem. 2002;277(28):25715–25721. [PubMed: 11994301]
150.
Buensuceso CS, Obergfell A, Soriani A. et al. Regulation of outside-in signaling in platelets by integrin-associated protein kinase C beta. J Biol Chem. 2005;280(1):644–653. [PubMed: 15536078]
151.
Kiosses WB, Shattil SJ, Pampori N. et al. Rac recruits high-affinity integrin alphavbeta3 to lamellipodia in endothelial cell migration. Nat Cell Biol. 2001;3(3):316–320. [PubMed: 11231584]
152.
Lee J, Jacobson K. The composition and dynamics of cell-substratum adhesions in locomoting fish keratocytes. J Cell Sci. 1997;110:2833–2844. [PubMed: 9427291]
153.
Roberts M, Barry S, Woods A. et al. PDGF-regulated rab4-dependent recycling of alphavbeta3 integrin from early endosomes is necessary for cell adhesion and spreading. Curr Biol. 2001;11(18):1392–1402. [PubMed: 11566097]
154.
Tadokoro S, Shattil SJ, Eto K. et al. Talin binding to integrin beta tails: A final common step in integrin activation. Science. 2003;302:103–106. [PubMed: 14526080]
155.
Calderwood DA. Talin controls integrin activation. Biochem Soc Trans. 2004;32:434–437. [PubMed: 15157154]
156.
Garcia-Alvarez B, de PeredaJM, Calderwood DA. et al. Structural determinants of integrin recognition by talin. Cell. 2003;11(1):49–58. [PubMed: 12535520]
157.
Yan B, Calderwood DA, Yaspan B. et al. Calpain cleavage promotes talin binding to the beta 3 integrin cytoplasmic domain. J Biol Chem. 2001;276(30):28164–28170. [PubMed: 11382782]
158.
Cuevas BD, Abell AN, Witowsky JA. et al. MEKK1 regulates calpain-dependent proteolysis of focal adhesion proteins for rear-end detachment of migrating fibroblasts. EMBO J. 2003;22(13):3346–3355. [PMC free article: PMC165646] [PubMed: 12839996]
159.
Franco SJ, Rodgers MA, Perrin BJ. et al. Calpain-mediated proteolysis of talin regulates adhesion dynamics. Nat Cell Biol. 2004;6(10):977–983. [PubMed: 15448700]
160.
Isenberg G, Goldmann WH. Peptide-specific antibodies localize the major lipid binding sites of talin dimers to oppositely arranged N-terminal 47 kDa subdomains. FEBS Lett. 1998;426:165–170. [PubMed: 9599000]
161.
Martel V, Racaud-Sultan C, Dupe S. et al. Conformation, localization, and integrin binding of talin depend on its interaction with phosphoinositides. J Biol Chem. 2001;276(24):21217–21227. [PubMed: 11279249]
162.
Di PaoloG, Pellegrini L, Letinic K. et al. Recruitment and regulation of phosphatidylinositol phosphate kinase type 1 gamma by the FERM domain of talin. Nature. 2002;420(6911):85–89. [PubMed: 12422219]
163.
Ling K, Doughman RL, Firestone AJ. et al. Type I gamma phosphatidylinositol phosphate kinase targets and regulates focal adhesions. Nature. 2002;420(6911):89–93. [PubMed: 12422220]
164.
Yin HL, Janmey PA. Phosphoinositide regulation of the actin cytoskeleton. Annu Rev Physiol. 2003;65:761–789. [PubMed: 12471164]
165.
Koo LY, Irvine DJ, Mayes AM. et al. Coregulation of cell adhesion by nanoscale RGD organization and mechanical stimulus. J Cell Sci. 2002;115(Pt 7):1423–1433. [PubMed: 11896190]
166.
Coussen F, Choquet D, Sheetz MP. et al. Trimers of the fibronectin cell adhesion domain localize to actin filament bundles and undergo rearward translocation. J Cell Sci. 2002;115(Pt 12):2581–2590. [PubMed: 12045228]
167.
Balaban NQ, Schwarz US, Riveline D. et al. Force and focal adhesion assembly: A close relationship studied using elastic micropatterned substrates. Nat Cell Biol. 2001;3(5):466–472. [PubMed: 11331874]
168.
Massia SP, Hubbell JA. An RGD spacing of 440 nm is sufficient for integrin alpha V beta 3-mediated fibroblast spreading and 140 nm for focal contact and stress fiber formation. J Cell Biol. 1991;114(5):1089–1100. [PMC free article: PMC2289117] [PubMed: 1714913]
169.
Arnold M, Cavalcanti-Adam EA, Glass R. et al. Activation of integrin function by nanopatterned adhesive interfaces. Chemphyschem. 2004;5(3):383–388. [PubMed: 15067875]
170.
Sawada Y, Sheetz MP. Force transduction by Triton cytoskeletons. J Cell Biol. 2002;156(4):609–615. [PMC free article: PMC2174068] [PubMed: 11839769]
171.
Wang HB, Dembo M, Hanks SK. et al. Focal adhesion kinase is involved in mechanosensing during fibroblast migration. Proc Natl Acad Sci USA. 2001;98(20):11295–11300. [PMC free article: PMC58723] [PubMed: 11572981]
172.
Trachslin J, Koch M, Chiquet M. Rapid and reversible regulation of collagen XII expression by changes in tensile stress. Exp Cell Res. 1999;247(2):320–328. [PubMed: 10066359]
173.
Chiquet M, Sarasa-Renedo A, Tunc-Civelek V. Induction of tenascin-C by cyclic tensile strain versus growth factors: Distinct contributions by Rho/ROCK and MAPK signaling pathways. Biochim Biophys Acta. 2004;1693(3):193–204. [PubMed: 15363633]
174.
Lo CM, Wang HB, Dembo M. et al. Cell movement is guided by the rigidity of the substrate. Biophys J. 2000;79(1):144–152. [PMC free article: PMC1300921] [PubMed: 10866943]
175.
Pelham JrRJ, Wang Y. Cell locomotion and focal adhesions are regulated by substrate flexibility. Proc Natl Acad Sci USA. 1997;94(25):13661–13665. [PMC free article: PMC28362] [PubMed: 9391082]
176.
Lehnert D, Wehrle-Haller B, David C. et al. Cell behavior on micropatterned substrates: Limits of extracellular matrix geometry for spreading and adhesion J Cell Sci 2003 . in press. [PubMed: 14657272]
177.
Duband JL, Nuckolls GH, Ishihara A. et al. Fibronectin receptor exhibits high lateral mobility in embryonic locomoting cells but is immobile in focal contacts and fibrillar streaks in stationary cells. J Cell Biol. 1988;107(4):1385–1396. [PMC free article: PMC2115255] [PubMed: 2971668]
178.
Axelrod D, Koppel DE, Schlessinger J. et al. Mobility measurement by analysis of fluorescence photobleaching recovery kinetics. Biophys J. 1976;16(9):1055–1069. [PMC free article: PMC1334945] [PubMed: 786399]
179.
White J, Stelzer E. Photobleaching GFP reveals protein dynamics inside live cells. Trends Cell Biol. 1999;9(2):61–65. [PubMed: 10087620]
180.
Danen EHJ, van RheenenJ, Franken W. et al. Integrins control motile strategy through a Rho-cofilin pathway. J Cell Biol. 2005;169(3):515–526. [PMC free article: PMC2171933] [PubMed: 15866889]
181.
Hinz B, Dugina V, Ballestrem C. et al. Alpha-smooth muscle actin is crucial for focal adhesion maturation in myofibroblasts. Mol Biol Cell. 2003;14(6):2508–2519. [PMC free article: PMC194898] [PubMed: 12808047]
182.
Tsuruta D, Gonzales M, Hopkinson SB. et al. Microfilament-dependent movement of the beta3 integrin subunit within focal contacts of endothelial cells. FASEB J. 2002;16(8):866–868. [PubMed: 11967230]
183.
Li R, Mitra N, Gratkowski H. et al. Activation of integrin alphaIIbbeta3 by modulation of transmembrane helix associations. Science. 2003;300(5620):795–798. [PubMed: 12730600]
184.
Evans RD, Perkins VC, Henry A. et al. A tumor-associated beta 1 integrin mutation that abrogates epithelial differentiation control. J Cell Biol. 2003;160(4):589–596. [PMC free article: PMC2173744] [PubMed: 12578911]
185.
Luo BH, Springer TA, Takagi J. Stabilizing the open conformation of the integrin headpiece with a glycan wedge increases affinity for ligand. Proc Natl Acad Sci USA. 2003;100:2403–2408. [PMC free article: PMC151353] [PubMed: 12604783]
186.
Luo BH, Carman CV, Takagi J. et al. Disrupting integrin transmembrane domain heterodimerization increases ligand binding affinity, not valency or clustering. Proc Natl Acad Sci USA. 2005;102(10):3679–3684. [PMC free article: PMC553322] [PubMed: 15738420]
187.
Litvinov RI, Nagaswami C, Vilaire G. et al. Functional and structural correlations of individual alphaIIb beta3 molecules. Blood. 2004;104(13):3979–3985. [PubMed: 15319287]
188.
Erb EM, Tangemann K, Bohrmann B. et al. Integrin alphaIIb beta3 reconstituted into lipid bilayers is nonclustered in its activated state but clusters after fibrinogen binding. Biochemistry. 1997;36(24):7395–7402. [PubMed: 9200686]
189.
DePasquale JA, Izzard CS. Accumulation of talin in nodes at the edge of the lamellipodium and separate incorporation into adhesion plaques at focal contacts in fibroblasts. J Cell Biol. 1991;113(6):1351–1359. [PMC free article: PMC2289043] [PubMed: 1904445]
190.
DeMali KA, Barlow CA, Burridge K. Recruitment of the Arp2/3 complex to vinculin: Coupling membrane protrusion to matrix adhesion. J Cell Biol. 2002;159(5):881–891. [PMC free article: PMC2173392] [PubMed: 12473693]
191.
Nayal A, Webb DJ, Horwitz AF. Talin: An emerging focal point of adhesion dynamics. Curr Opin Cell Biol. 2004;16(1):94–98. [PubMed: 15037311]
192.
Honda A, Nogami M, Yokozeki T. et al. Phosphatidylinositol 4-phosphate 5-kinase alpha is a downstream effector of the small G protein ARF6 in membrane ruffle formation. Cell. 1999;99(5):521–532. [PubMed: 10589680]
193.
Santy LC, Casanova JE. Activation of ARF6 by ARNO stimulates epithelial cell migration through downstream activation of both Rac1 and phospholipase D. J Cell Biol. 2001;154(3):599–610. [PMC free article: PMC2196419] [PubMed: 11481345]
194.
Radhakrishna H, Al-Awar O, Khachikian Z. et al. ARF6 requirement for Rac ruffling suggests a role for membrane trafficking in cortical actin rearrangements. J Cell Sci. 1999;112(Pt 6):855–866. [PubMed: 10036235]
195.
Fincham VJ, Frame MC. The catalytic activity of Src is dispensable for translocation to focal adhesions but controls the turnover of these structures during cell motility. EMBO J. 1998;17(1):81–92. [PMC free article: PMC1170360] [PubMed: 9427743]
196.
Frame MC, Fincham VJ, Carragher NO. et al. v-Src's hold over actin and cell adhesions. Nat Rev Mol Cell Biol. 2002;3(4):233–245. [PubMed: 11994743]
197.
Westhoff MA, Serrels B, Fincham VJ. et al. SRC-mediated phosphorylation of focal adhesion kinase couples actin and adhesion dynamics to survival signaling. Mol Cell Biol. 2004;24(18):8113–8133. [PMC free article: PMC515031] [PubMed: 15340073]
198.
Dourdin N, Bhatt AK, Dutt P. et al. Reduced cell migration and disruption of the actin cytoskeleton in calpain-deficient embryonic fibroblasts. J Biol Chem. 2001;276(51):48382–48388. [PubMed: 11602605]
199.
Carragher NO, Frame MC. Calpain: A role in cell transformation and migration. Int J Biochem Cell Biol. 2002;34(12):1539–1543. [PubMed: 12379276]
200.
Huttenlocher A, Palecek SP, Lu Q. et al. Regulation of cell migration by the calcium-dependent protease calpain. J Biol Chem. 1997;272(52):32719–32722. [PubMed: 9407041]
201.
Bretscher MS, Aguado-Velasco C. Membrane traffic during cell locomotion. Curr Opin Cell Biol. 1998;10(4):537–541. [PubMed: 9719876]
202.
Woods AJ, White DP, Caswell PT. et al. PKD1/PKCmicro promotes alphavbeta3 integrin recycling and delivery to nascent focal adhesions. EMBO J. 2004;23(13):2531–2543. [PMC free article: PMC449777] [PubMed: 15192707]
203.
Roberts MS, Woods AJ, Dale TC. et al. Protein kinase B/Akt acts via glycogen synthase kinase 3 to regulate recycling of alpha v beta 3 and alpha 5 beta 1 integrins. Mol Cell Biol. 2004;24(4):1505–1515. [PMC free article: PMC344170] [PubMed: 14749368]
204.
Zhang H, Berg JS, Li Z. et al. Myosin-X provides a motor-based link between integrins and the cytoskeleton. Nat Cell Biol. 2004;6(6):523–531. [PubMed: 15156152]
Copyright © 2000-2013, Landes Bioscience.
Bookshelf ID: NBK6613

Views

  • PubReader
  • Print View
  • Cite this Page

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...