首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
Kindlins are essential for integrin activation in cell systems and do so by working in a cooperative fashion with talin via their direct interaction with integrin β cytoplasmic tails (CTs). Kindlins interact with the membrane-distal NxxY motif, which is distinct from the talin-binding site within the membrane-proximal NxxY motif. The Tyr residues in both motifs can be phosphorylated, and it has been suggested that this modification of the membrane-proximal NxxY motif negatively regulates interaction with the talin head domain. However, the influence of Tyr phosphorylation of the membrane-distal NxxY motif on kindlin binding is unknown. Using mutational analyses and phosphorylated peptides, we show that phosphorylation of the membrane-distal NITY759 motif in the β3 CT disrupts kindlin-2 recognition. Phosphorylation of this membrane-distal Tyr also disables the ability of kindlin-2 to coactivate the integrin. In direct binding studies, peptides corresponding to the non-phosphorylated β3 CT interacted well with kindlin-2, whereas the Tyr759-phosphorylated peptide failed to bind kindlin-2 with measurable affinity. These observations indicate that transitions between the phosphorylated and non-phosphorylated states of the integrin β3 CT determine reactivity with kindlin-2 and govern the role of kindlin-2 in regulating integrin activation.  相似文献   

2.
Heterotrimeric G protein Gα13 is known to transmit G protein–coupled receptor (GPCR) signals leading to activation of RhoA and plays a role in cell migration. The mechanism underlying the role of Gα13 in cell migration, however, remains unclear. Recently we found that Gα13 interacts with the cytoplasmic domain of integrin β3 subunits in platelets via a conserved ExE motif. Here we show that a similar direct interaction between Gα13 and the cytoplasmic domain of the integrin β1 subunit plays a critical role in β1-dependent cell migration. Point mutation of either glutamic acid in the Gα13-binding 767EKE motif in β1 or treatment with a peptide derived from the Gα13-binding sequence of β1 abolished Gα13–β1 interaction and inhibited β1 integrin–dependent cell spreading and migration. We further show that the Gα131 interaction mediates β1 integrin–dependent Src activation and transient RhoA inhibition during initial cell adhesion, which is in contrast to the role of Gα13 in mediating GPCR-dependent RhoA activation. These data indicate that Gα13 plays dynamic roles in both stimulating RhoA via a GPCR pathway and inhibiting RhoA via an integrin signaling pathway. This dynamic regulation of RhoA activity is critical for cell migration on β1 integrin ligands.  相似文献   

3.
We have used gene disruption to isolate two talin (−/−) ES cell mutants that contain no intact talin. The undifferentiated cells (a) were unable to spread on gelatin or laminin and grew as rounded colonies, although they were able to spread on fibronectin (b) showed reduced adhesion to laminin, but not fibronectin (c) expressed much reduced levels of β1 integrin, although levels of α5 and αV were wild-type (d) were less polarized with increased membrane protrusions compared with a vinculin (−/−) ES cell mutant (e) were unable to assemble vinculin or paxillin-containing focal adhesions or actin stress fibers on fibronectin, whereas vinculin (−/−) ES cells were able to assemble talin-containing focal adhesions. Both talin (−/−) ES cell mutants formed embryoid bodies, but differentiation was restricted to two morphologically distinct cell types. Interestingly, these differentiated talin (−/−) ES cells were able to spread and form focal adhesion-like structures containing vinculin and paxillin on fibronectin. Moreover, the levels of the β1 integrin subunit were comparable to those in wild-type ES cells. We conclude that talin is essential for β1 integrin expression and focal adhesion assembly in undifferentiated ES cells, but that a subset of differentiated cells are talin independent for both characteristics.  相似文献   

4.
The contributions of integrins to cellular responses depend upon their activation, which is regulated by binding of proteins to their cytoplasmic tails. Kindlins are integrin cytoplasmic tail binding partners and are essential for optimal integrin activation, and kindlin-3 fulfills this role in hematopoietic cells. Here, we used human platelets and human erythroleukemia (HEL) cells, which express integrin αIIbβ3, to investigate whether phosphorylation of kindlin-3 regulates integrin activation. When HEL cells were stimulated with thrombopoietin or phorbol 12-myristate 13-acetate (PMA), αIIbβ3 became activated as evidenced by binding of an activation-specific monoclonal antibody and soluble fibrinogen, adherence and spreading on fibrinogen, colocalization of β3 integrin and kindlin-3 in focal adhesions, and enhanced β3 integrin-kindlin-3 association in immunoprecipitates. Kindlin-3 knockdown impaired adhesion and spreading on fibrinogen. Stimulation of HEL cells with agonists significantly increased kindlin-3 phosphorylation as detected by mass spectrometric sequencing. Thr482 or Ser484 was identified as a phosphorylation site, which resides in a sequence not conserved in kindlin-1 or kindlin-2. These same residues were phosphorylated in kindlin-3 when platelets were stimulated with thrombin. When expressed in HEL cells, T482A/S484A kindlin-3 decreased soluble ligand binding and cell spreading on fibrinogen compared with wild-type kindlin-3. A membrane-permeable peptide containing residues 476–485 of kindlin-3 was introduced into HEL cells and platelets; adhesion and spreading of both cell types were blunted compared with a scrambled control peptide. These data identify a role of kindlin-3 phosphorylation in integrin β3 activation and provide a basis for functional differences between kindlin-3 and the two other kindlin paralogs.  相似文献   

5.
Cell migration requires the coordination of adhesion site assembly and turnover. Canonical models for nascent adhesion formation postulate that integrin binding to extracellular matrix (ECM) proteins results in the rapid recruitment of cytoskeletal proteins such as talin and paxillin to integrin cytoplasmic domains. It is thought that integrin-talin clusters recruit and activate tyrosine kinases such as focal adhesion kinase (FAK). However, the molecular connections of this linkage remain unresolved. Our recent findings support an alternative model whereby FAK recruits talin to new sites of β1 integrin-mediated adhesion in mouse embryonic fibroblasts and human ovarian carcinoma cells. This is dependent on a direct binding interaction between FAK and talin and occurs independently of direct talin binding to β1 integrin. Herein, we discuss differences between nascent and mature adhesions, interactions between FAK, talin and paxillin, possible mechanisms of FAK activation and how this FAK-talin complex may function to promote cell motility through increased adhesion turnover.  相似文献   

6.
Integrin heterodimers acquire high affinity to endothelial ligands by extensive conformational changes in both their α and β subunits. These heterodimers are maintained in an inactive state by inter-subunit constraints. Changes in the cytoplasmic interface of the integrin heterodimer (referred to as inside-out integrin activation) can only partially remove these constraints. Full integrin activation is achieved when both inter-subunit constraints and proper rearrangements of the integrin headpiece by its extracellular ligand (outside-in activation) are temporally coupled. A universal regulator of these integrin rearrangements is talin1, a key integrin-actin adaptor that regulates integrin conformation and anchors ligand-occupied integrins to the cortical cytoskeleton. The arrest of rolling leukocytes at target vascular sites depends on rapid activation of their α4 and β2 integrins at endothelial contacts by chemokines displayed on the endothelial surface. These chemotactic cytokines can signal within milliseconds through specialized Gi-protein coupled receptors (GPCRs) and Gi-triggered GTPases on the responding leukocytes. Some chemokine signals can alter integrin conformation by releasing constraints on integrin extension, while other chemokines activate integrins to undergo conformational activation mainly after ligand binding. Both of these modalities involve talin1 activation. In this opinion article, I propose that distinct chemokine signals induce variable strengths of associations between talin1 and different target integrins. Weak interactions of the integrin cytoplasmic tail with talin1 (the cytoplasmic integrin ligand) dissociate unless the extracellular ligand can simultaneously occupy the integrin headpiece and transmit, within milliseconds, proper allosteric changes across the integrin heterodimer back to the tail-talin1 complex. The fate of this bi-directional occupancy of integrins by both their extracellular and intracellular ligands is likely to benefit from immobilization of both ligands to cortical cytoskeletal elements. To properly anchor talin1 onto the integrin tail, a second integrin partner, Kindlin-3 may be also required, although an evidence that both partners can simultaneously bind the same integrin heterodimer is still missing. Once linked to the cortical actin cytoskeleton, the multi-occupied integrin complex can load weak forces, which deliver additional allosteric changes to the integrin headpiece resulting in further bond strengthening. Surface immobilized chemokines are superior to their soluble counterparts in driving this bi-directional occupancy process, presumably due to their ability to facilitate local co-occupancy of individual integrin heterodimers with talin1, Kindlin-3 and surface-bound extracellular ligands.Key words: adhesion, migration, endothelium, cytoskeleton, shear stress, immunityFirm adhesion of leukocytes to blood vessels is tightly regulated by integrins and their cognate ligands.1,2 These include the α4 integrins, VLA-4 (α4β1) and α4β7, and the β2 integrins, LFA-1 (αLβ2) and Mac-1 (αMβ2). Accumulated data suggest that these counter-receptors are structurally adapted to operate under disruptive blood-derived shear forces.3 A remarkable feature of leukocyte integrins is that their affinity state and microclustering can be regulated within fractions of seconds.4,5 The most robust signals for leukocyte integrins are transduced by chemoattractants, mostly chemokines displayed on the vessel wall.6 A growing body of evidence suggests that the Gi protein coupled receptors of these endothelial chemokines elicit diverse signaling pathways in distinct leukocyte subtypes,2,22 which use two common downstream elements to coactive all leukocyte integrins: talin1 and Kindlin-3.7 In this review, I will describe a model explaining how chemokine signals to these elements regulate the conformation of all leukocyte integrins by facilitating a coupled bi-directional occupancy and activation via both their cytoplasmic and headpiece domains.Recent structural and biophysical studies suggest that leukocyte integrins can alternate between inactive bent conformers, which are clasped heterodimers, and variable unclasped heterodimers with extended ectodomains exhibiting intermediate and high affinity to ligand.5 Most leukocyte integrins are maintained in an inactive resting state,2 whereas in situ chemokine-stimulated integrins unfold and extend 10–25 nm above the cell surface, allowing their headpiece to readily recognize immobilized ligand on a counter surface.8 These extended integrins must undergo extensive rearrangements of their headpiece I-domains induced by external endothelial-displayed ligands in order to arrest rolling leukocytes on blood vessel walls. In leukocytes, these two canonical switches are very short-lived, implying the necessity for a stabilization. It is therefore likely that any type of robust integrin activation must involve bi-directional occupancy of the integrin by both its extracellular ligand and one or more cytoplasmic partners.9The main cytoplasmic integrin-activating adaptor in leukocytes and platelets is talin1.10,11 Talin knock down in multiple cell types results in nearly total loss of integrin activation.12,13 This actin binding adaptor binds different integrin β subunit tails with low affinity,14 which can be locally increased by in situ generated PI(4,5)P2 (PIP2). This phosphoinositide presumably binds to the FERM domain within the talin head and thereby enhances talin binding to a membrane proximal NPXY motif on the β integrin tail, a key event in integrin heterodimer unclasping.15,16 Recent studies suggest, however, that mere talin association may be insufficient to unclasp and activate the integrin heterodimer. Thus, the beta subunit tail may need to get co-occupied by the integrin co-activator, Kindlin, in order to optimize talin association with this integrin subunit.17,18 In leukocytes, Talin1 and the Kindlin family member, Kindlin 3 co-activate both VLA-4 and LFA-1 and this co-activation is dramatically enhanced by multiple chemokine triggered effectors, the nature of which has begun to unfold19 (Fig. 1). I would like to propose that talin1-Kindlin-3 co-binding to the β tails of these and other leukocyte integrins is insufficient to switch these integrins to a conformation able to bind their soluble extracellular ligands due to fast dissocia-tion of PIP2-activated talin1 from the integrin cytoplasmic tail complex. This short lived talin-integrin complex may, on the other hand, get stabilized, if the integrin headpiece can simultaneously bind an immobilized extracellular ligand and undergo immediate outside-in activation, before the talin1 has dissociated from the integrin beta tail (Fig. 1). Such full confor-mational switch can result in additional allosteric changes in the integrin-bound talin which may expose vinculin binding sites and further increase talin-actin associations that reinforce this bi-directional allosteric integrin activation.20Open in a separate windowFigure 1Bi-directional integrin activation requires simultaneous co-occupancy of the integrin heterodimer by extracellular and cytoplasmic ligands. A proposed scheme for chemokine-triggered integrin activation on leukocytes. Integrin conformation is allosterically modulated in a bidirectional manner by at least two sets of ligands, extracellular and cytoplasmic. The degree of activation is dictated primarily by unclasping of the integrin heterodimer, a process dependent on the binding of the activated talin FERM domain to a specific site on the integrin β tail. (1) Inactive integrin. (2–5) Four postulated integrin conformations triggered by distinct chemokine signals. (2) Talin FERM domain activation close to the target integrin is a rate limiting step in integrin activation. This activation is triggered by PIP2 locally generated by talin-associated PIP5Kγ (purple rectangle) stimulated by a nearby Gi-coupled chemokine receptor. (3) Kindlin-3 binding to the integrin β tail stabilizes the otherwise weak talin1-integrin tail complex. The activated integrin can bind a soluble extracellular ligand with a low affinity due to a high koff of the soluble ligand from the integrin headpiece. (4) In the absence of Kindlin-3, chemokine triggered, PIP2-activated talin1 binds only transiently the integrin tail (High koff). The semiactivated integrin, even if occupied by an immobilized extracellular ligand, cannot undergo full bi-directional activation. (5) When both the extracellular ligand and talin are properly anchored, their escape from the integrin is dramatically reduced, lowering the koff. Low koff increases the probability of simultaneous bi-directional occupancy of both the integrin headpiece by the extracellular ligand and of the integrin tail by talin1 and Kindlin-3. This results in optimal bi-directional integrin activation and unclasping of the heterodimer. Stable linkages also allow this bi-directionally occupied integrin to undergo extensive mechanical strengthening by low forces applied on the headpiece; this further activates the headpiece I domains, further separates the β and α subunits from each other, and maximally stabilizes the unclasped integrin. Force application through the high affinity-talin complex can stretch the talin rod domain and expose vinculin binding sites (VBS). Since integrin ligands are generally multivalent, rapid integrin dimerization can take place to further stabilize the focal adhesion (not shown). Additional cytoplasmic partners of specific leukocyte integrins like a-actinin, L-plastin and RAPL may further strengthen subsets of focal adhesions. These and other cytosolic partners bind different integrin targets with different affinities. Therefore the effect of each of these partners on both the kinetics and stability of the talin1-integrin tail complex may vary with the cell type, the integrin type, the strength of the chemokine signal and the proximity between the integrin and its stimulatory GPCR.How can such postulated simultaneous bi-directional occupancy of a leukocyte integrin can be so rapidly triggered by a chemokine signal encountered during leukocyte rolling on blood vessels? An attractive mechanism for in situ facilitation of talin1 binding to the integrin β tail by chemokine signals involves chemokine triggered Gi stimulated RhoA and Rac1 GTPases and their downstream target, the PIP2 generating enzyme PIP5Kγ in the vicinity of the in situ activated integrin19 (Fig. 1). Additional talin1 molecules may also be recruited to the vicinity of this initially stimulated integrin by RIAM,21 an effector that associates with activated Rap-1, one of the key chemokine stimulated GTPases involved in rapid integrin mediated activation in both leukocytes and platelets.22,23 To bidirectionally bind and activate their integrin targets, both the cytoplasmic integrin ligands, Talin1 and Kindlin-3 and the extracellular integrin ligand may need to achieve low dissociation rates from the integrin tail and headpiece, respectively. Why would an immobilized extracellular ligand be superior to soluble extracellular ligand in capacity to bi-directionally bind and activate a leukocyte integrin? The probability that a given surface-bound ligand, rather than a soluble integrin ligand would escape from its cognate integrin receptor following its dissociation is very small, since reactants in viscous medium are more likely to recombine than to diffuse apart.24 Thus, surface-immobilized single integrin ligands may rebind the integrins they recenty dissociated from much more frequently than their soluble counterparts. Similarly, the cytoplasmic ligands talin1 and Kindlin may need to remain immobile once occupying their target integrin tail. Such immobilization of talin1 can be optimized by talin anchoring to the cortical cytoskeleton.25 Talin may be also restricted from immediate dissociation from the integrin tail by Kindlin-3. An optimal integrin activating chemokine signal would therefore not only need to recruit and induce talin1 association with the β subunit of the target integrin and opening of the integrin clasp, but also need to keep the talin in an immobile state, and thereby maintain its low dissociation rate from its integrin tail sites.As both the integrin headpiece and the integrin subunits are predicted to undergo faster opening and separation in the presence of applied forces,26,27 another tradeoff of this postulated immobilization of both the intracellular and extracellular integrin ligands is optimal force sensing of the integrin heterodimer. Application of force on the bidirectionally occupied integrin and its cognate ligands would be possible only if the integrin, its extracellular ligand, and talin1 are all properly anchored.3,28,29 Force transduction through the integrin-talin1 complex can transmit additional conformational changes on the integrin-occupied talin by exposing vinculin binding sites on the talin rod.30 Additional chemokine signals may induce talin rod phosphorylation and other changes in actin-talin associations (Fig. 1) that may further facilitate talin anchorage to the cortical cytoskeleton and subsequent microclustering of adjacent ligand-occupied integrins. It is well recognized that ligand occupancy anchors integrins to the cortical cytoskeleton.31 Thus, the anchorage states of both the extracellular and the cytoplasmic ligands of a given integrin may facilitate bidirectional integrin occupancy and optimize force driven bi-directional activation of the integrin-ligand complex and subsequent dimerization of ligand-occupied integrins. The ability of different integrin-ligand complexes to undergo diverse mechanochemical rearrangements provides a broad spectrum of integrin-ligand bond strengths, accounting for the unique capacity of chemokine stimulated leukocyte integrins to support both firm and reversible adhesions of leukocytes to their endothelial ligands.  相似文献   

7.
The αvβ3 integrin participates in cell morphogenesis, growth factor signaling, and cell survival. Activation of the integrin is central to these processes and is influenced by specific ECM components, which engage both integrins and syndecans. This paper demonstrates that the αvβ3 integrin and syndecan-1 (S1) are functionally coupled. The integrin is dependent on the syndecan to become activated and to mediate signals required for MDA-MB-231 and MDA-MB-435 human mammary carcinoma cell spreading on vitronectin or S1-specific antibody. Coupling of the syndecan to αvβ3 requires the S1 ectodomain (ED), as ectopic expression of glycosylphosphatidylinositol-linked S1ED enhances αvβ3 recognition of vitronectin; and treatments that target this domain, including competition with recombinant S1ED protein or anti-S1ED antibodies, mutation of the S1ED, or down-regulation of S1 expression by small-interfering RNAs, disrupt αvβ3-dependent cell spreading and migration. Thus, S1 is likely to be a critical regulator of many cellular behaviors that depend on activated αvβ3 integrins.  相似文献   

8.
Studying the tight activity regulation of platelet-specific integrin αIIbβ3 is foundational and paramount to our understanding of integrin structure and activation. αIIbβ3 is essential for the aggregation and adhesion function of platelets in hemostasis and thrombosis. Structural and mutagenesis studies have previously revealed the critical role of αIIbβ3 transmembrane (TM) association in maintaining the inactive state. Gain-of-function TM mutations were identified and shown to destabilize the TM association leading to integrin activation. Studies using isolated TM peptides have suggested an altered membrane embedding of the β3 TM α-helix coupled with αIIbβ3 activation. However, controversies remain as to whether and how the TM α-helices change their topologies in the context of full-length integrin in native cell membrane. In this study, we utilized proline scanning mutagenesis and cysteine scanning accessibility assays to analyze the structure and function correlation of the αIIbβ3 TM domain. Our identification of loss-of-function proline mutations in the TM domain suggests the requirement of a continuous TM α-helical structure in transmitting activation signals bidirectionally across the cell membrane, characterized by the inside-out activation for ligand binding and the outside-in signaling for cell spreading. Similar results were found for αLβ2 and α5β1 TM domains, suggesting a generalizable mechanism. We also detected a topology change of β3 TM α-helix within the cell membrane, but only under conditions of cell adhesion and the absence of αIIb association. Our data demonstrate the importance of studying the structure and function of the integrin TM domain in the native cell membrane.  相似文献   

9.
Understanding how cells integrate multiple signaling pathways to achieve specific cell differentiation is a challenging question in cell biology. We have explored the physiological presentation of BMP-2 by using a biomaterial that harbors tunable mechanical properties to promote localized BMP-2 signaling. We show that matrix-bound BMP-2 is sufficient to induce β3 integrin–dependent C2C12 cell spreading by overriding the soft signal of the biomaterial and impacting actin organization and adhesion site dynamics. In turn, αvβ3 integrin is required to mediate BMP-2–induced Smad signaling through a Cdc42–Src–FAK–ILK pathway. β3 integrin regulates a multistep process to control first BMP-2 receptor activity and second the inhibitory role of GSK3 on Smad signaling. Overall, our results show that BMP receptors and β3 integrin work together to control Smad signaling and tensional homeostasis, thereby coupling cell adhesion and fate commitment, two fundamental aspects of developmental biology and regenerative medicine.  相似文献   

10.
Osteoclast interaction with extracellular matrix drives the sequential events that end with bone resorption. However, the role of matrix proteins is not yet fully understood. We studied this problem on human osteoclast-like cells derived from giant cell tumors of bone (GCT cells). On GCT cells we considered cytoskeletal organization, adhesion properties, and integrin expression upon plating in serum-free medium onto fibronectin (FN), collagen (COL), thrombospondin (TSP), bone sialoprotein (BSPII), and osteopontin (OPN). GCT cells promptly adhered and spread on FN, BSPII, and OPN, while only 50% adhered on COL and none on TSP. The integrin β1 chain was always associated to focal adhesions, while the αvβ3 heterodimer was detected in focal contacts only upon plating on BSPII, OPN, and FN. The focal clustering of β1 was impaired by monensin treatment, indicating that endogenous FN secretion was required to drive β1 into focal contacts. Conversely, αvβ3 clustering was also not affected by monensin when cells were plated onto plasma FN. Immunoprecipitation of metabolically labeled GCT cell lysates showed that three different heterodimers (αvβ3, α3β1, and α5β1) were assembled. Adhesion to FN was completely inhibited by β1 antibodies at dilutions up to 1:400, while β3 antibodies, at similar dilutions, impaired spreading but not adhesion. We conclude that αvβ33 is the main integrin used by GCT cells in bone recognition. We also suggest that selected substrata may induce the release and the organization of endogenous FN that eventually drives the recruitment of a β1 integrin receptor into focal contacts.  相似文献   

11.
Binding of integrins to ligands provides anchorage and signals for the cell, making them prime candidates for mechanosensing molecules. How force regulates integrin–ligand dissociation is unclear. We used atomic force microscopy to measure the force-dependent lifetimes of single bonds between a fibronectin fragment and an integrin α5β1-Fc fusion protein or membrane α5β1. Force prolonged bond lifetimes in the 10–30-pN range, a counterintuitive behavior called catch bonds. Changing cations from Ca2+/Mg2+ to Mg2+/EGTA and to Mn2+ caused longer lifetime in the same 10–30-pN catch bond region. A truncated α5β1 construct containing the headpiece but not the legs formed longer-lived catch bonds that were not affected by cation changes at forces <30 pN. Binding of monoclonal antibodies that induce the active conformation of the integrin headpiece shifted catch bonds to a lower force range. Thus, catch bond formation appears to involve force-assisted activation of the headpiece but not integrin extension.  相似文献   

12.
The interaction of β1 integrin receptors and different extracellular matrix molecules during neuronal development was investigated by comparing both migration and morphological differentiation of D3 wild-type embryonic stem (ES) cell line-derived neural precursor cells with those of the β1 integrin knockout ES cell line G201. Analysing neurosphere explants on laminin and fibronectin as major β1 integrin ligands, the maximal spreading of outward migrating neuronal cells was determined. Compared with gelatine as a standard substrate, migration was found to be significantly increased for D3-derived neurospheres on fibronectin and laminin-1. These matrix effects were found to be even enhanced for G201 preparations. In addition, also the differentiation of wild-type and β1 integrin −/− neurones – as determined by MAP-2- and HNK-1-immunoreactive processes – was found to be increased on fibronectin and laminin when compared to gelatine standards. In the respective knockout preparations on these matrices, again perturbation effects were less pronounced than on gelatine. Our observations indicate that laminin and fibronectin are involved both in β1 integrin-dependent and -independent signalling mechanisms during neurogenesis. Upregulation of compensatory mechanisms such as β1 integrin-independent receptors for laminin and fibronectin might be responsible for the much less pronounced perturbations of G201 neural precursor migration and differentiation on these two substrates than on gelatine.  相似文献   

13.
According to previous research, integrin β1 and ILK play an important role in the extracellular matrix (ECM)–integrin–cytoskeleton pathway for mechanotransduction. The aim of this study was to investigate strain induced integrin β1 and ILK expression in three-dimensional (3D) and in two-dimensional (2D) cultured rat skeletal myoblasts. Sprague–Dawley (SD) rat skeletal myoblasts were isolated and seeded on the PLGA-collagen composite scaffolds. The 3D cultured and the conventionally 2D monolayer cultured myoblasts were loaded 2000 μstrain tensile strain at 0.5 Hz for 2 h, 4 h, 8 h, 12 h and 24 h, respectively with the self-made four-point bending system. The expressions of integrin β1 and ILK mRNA were measured by RT-PCR and the different changes between the 3D and 2D cultures were compared. The mRNA expression levels of both integrin β1 and ILK were up regulated after mechanical loading (P < 0.05), meanwhile, it was higher and peaked faster in 3D cultures than in the 2D cultures. It can be concluded that the ECM–integrin–cytoskeleton pathway responds to tensile strain by elevated expression of integrin β1 and ILK, and the response is stronger in 3D cultures than in conventional 2D monolayer cultures.  相似文献   

14.
Integrins are bidirectional, allosteric transmembrane receptors that play a central role in hemostasis and arterial thrombosis. Using cryo-electron microscopy, multireference single-particle reconstruction methods, and statistics-based computational fitting approaches, we determined three-dimensional structures of human integrin αIIbβ3 embedded in a lipid bilayer (nanodiscs) while bound to domains of the cytosolic regulator talin and to extracellular ligands. We also determined the conformations of integrin in solution by itself to localize the membrane and the talin-binding site. To our knowledge, our data provide unprecedented three-dimensional information about the conformational states of intact, full-length integrin within membrane bilayers under near-physiological conditions and in the presence of cytosolic activators and extracellular ligands. We show that αIIbβ3 integrins exist in a conformational equilibrium clustered around four main states. These conformations range from a compact bent nodule to two partially extended intermediate conformers and finally to a fully upright state. In the presence of nanodiscs and the two ligands, the equilibrium is significantly shifted toward the upright conformation. In this conformation, the receptor extends ∼20 nm upward from the membrane. There are no observable contacts between the two subunits other than those in the headpiece near the ligand-binding pocket, and the α- and β-subunits are well separated with their cytoplasmic tails ∼8 nm apart. Our results indicate that extension of the ectodomain is possible without separating the legs or extending the hybrid domain, and that the ligand-binding pocket is not occluded by the membrane in any conformations of the equilibrium. Further, they suggest that integrin activation may be influenced by equilibrium shifts.  相似文献   

15.
Integrins are heterodimeric transmembrane receptors that physically link the extracellular matrix (ECM) to the intracellular actin cytoskeleton, and are also signaling molecules that transduce signals bi-directionally across the plasma membrane. Integrin regulation is essential for tumor cell migration in response to growth factors. c-Abl kinase is a nonreceptor tyrosine kinase and is critical for signaling transduction from various receptors. Here we show that c-Abl kinase is involved in A375 cell migration mediated by αvβ3 integrin in response to PDGF stimulation. c-Abl kinase colocalizes with αvβ3 integrin dynamically and affects αvβ3 integrin affinity by regulating its cluster. The interaction between c-Abl kinase and αvβ3 integrin was dependent on the activity of c-Abl kinase induced by PDGF stimulation, but was not dependent on the binding of αvβ3 integrin with its ligands, suggesting that c-Abl kinase is not involved in the outside-in signaling of αvβ3 integrin. Talin head domain was required for the interaction between c-Abl kinase and αvβ3 integrin, and the SH3 domain of c-Abl kinase was involved in its interaction with talin and αvβ3 integrin. Taken together, we have uncovered a novel and critical role of c-Abl kinase in αvβ3 integrin mediated melanoma cell migration.  相似文献   

16.
Integrins are heterodimeric (αβ) cell surface receptors that are activated to a high affinity state by the formation of a complex involving the α/β integrin transmembrane helix dimer, the head domain of talin (a cytoplasmic protein that links integrins to actin), and the membrane. The talin head domain contains four sub-domains (F0, F1, F2 and F3) with a long cationic loop inserted in the F1 domain. Here, we model the binding and interactions of the complete talin head domain with a phospholipid bilayer, using multiscale molecular dynamics simulations. The role of the inserted F1 loop, which is missing from the crystal structure of the talin head, PDB:3IVF, is explored. The results show that the talin head domain binds to the membrane predominantly via cationic regions on the F2 and F3 subdomains and the F1 loop. Upon binding, the intact talin head adopts a novel V-shaped conformation which optimizes its interactions with the membrane. Simulations of the complex of talin with the integrin α/β TM helix dimer in a membrane, show how this complex promotes a rearrangement, and eventual dissociation of, the integrin α and β transmembrane helices. A model for the talin-mediated integrin activation is proposed which describes how the mutual interplay of interactions between transmembrane helices, the cytoplasmic talin protein, and the lipid bilayer promotes integrin inside-out activation.  相似文献   

17.
18.
Regulation of vascular homeostasis depends upon collaboration between cells of the vessel wall and blood coagulation system. A direct interaction between integrin αVβ3 on endothelial cells and smooth muscle cells and prothrombin, the pivotal proenzyme of the blood coagulation system, is demonstrated and activation of the integrin is required for receptor engagement. Evidence that prothrombin is a ligand for αVβ3 on these cells include: (a) prothrombin binds to purified αVβ3 via a RGD recognition specificity; (b) prothrombin supports αVβ3-mediated adhesion of stimulated endothelial cells and smooth muscle cells; and (c) endothelial cells, either in suspension and in a monolayer, recognize soluble prothrombin via αVβ3. αVβ3-mediated cell adhesion to prothrombin, but not to fibrinogen, required activation of the receptor. Thus, the functionality of the αVβ3 receptor is ligand defined, and prothrombin and fibrinogen represent activation- dependent and activation-independent ligands. Activation of αVβ3 could be induced not only by model agonists, PMA and Mn2+, but also by a physiologically relevant agonist, ADP. Inhibition of protein kinase C and calpain prevented activation of αVβ3 on vascular cells, suggesting that these molecules are involved in the inside-out signaling events that activate the integrin. The capacity of αVβ3 to interact with prothrombin may play a significant role in the maintenance of hemostasis; and, at a general level, ligand selection by αVβ3 may be controlled by the activation state of this integrin.  相似文献   

19.
Integrin-dependent adhesion sites consist of clustered integrins that transmit mechanical forces and provide signaling required for cell survival and morphogenesis. Despite their importance, the regulation of integrin clustering by the cytoplasmic adapter protein talin (Tal) and phosphatidylinositol (PI)-4,5-biphosphate (PI(4,5)P2) lipids nor their dynamic coupling to the actin cytoskeleton is fully understood. By using a Tal-dependent integrin clustering assay in intact cells, we identified a PI(4,5)P2-binding basic ridge spanning across the F2 and F3 domains of the Tal head that regulates integrin clustering. Clustering requires a new PI(4,5)P2-binding site in F2 and is negatively regulated by autoinhibitory interactions between F3 and the Tal rod (Tal-R). The release of the Tal-R exposes a new β3-integrin–binding site in F3, enabling interaction with a membrane proximal acidic motif, which involves the formation of salt bridges between K316 and K324 with E726 and D723, respectively. This interaction shields the β-integrin tail from reassociation with its α subunit, thereby maintaining the integrin in a substrate-binding and clustering-competent form.  相似文献   

20.
The interaction of cells with fibronectin generates a series of complex signaling events that serve to regulate several aspects of cell behavior, including growth, differentiation, adhesion, and motility. The formation of a fibronectin matrix is a dynamic, cell-mediated process that involves both ligation of the α5β1 integrin with the Arg-Gly-Asp (RGD) sequence in fibronectin and binding of the amino terminus of fibronectin to cell surface receptors, termed “matrix assembly sites,” which mediate the assembly of soluble fibronectin into insoluble fibrils. Our data demonstrate that the amino-terminal type I repeats of fibronectin bind to the α5β1 integrin and support cell adhesion. Furthermore, the amino terminus of fibronectin modulates actin assembly, focal contact formation, tyrosine kinase activity, and cell migration. Amino-terminal fibronectin fragments and RGD peptides were able to cross-compete for binding to the α5β1 integrin, suggesting that these two domains of fibronectin cannot bind to the α5β1 integrin simultaneously. Cell adhesion to the amino-terminal domain of fibronectin was enhanced by cytochalasin D, suggesting that the ligand specificity of the α5β1 integrin is regulated by the cytoskeleton. These data suggest a new paradigm for integrin-mediated signaling, where distinct regions within one ligand can modulate outside-in signaling through the same integrin.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号