首页 | 本学科首页   官方微博 | 高级检索  
   检索      


Migfilin, a Molecular Switch in Regulation of Integrin Activation
Authors:Sujay Subbayya Ithychanda  Mitali Das  Yan-Qing Ma  Keyang Ding  Xiaoxia Wang  Sudhiranjan Gupta  Chuanyue Wu  Edward F Plow  and Jun Qin
Institution:Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195 and the §Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
Abstract:The linkage of heterodimeric (α/β) integrin receptors with their extracellular matrix ligands and intracellular actin cytoskeleton is a fundamental step for controlling cell adhesion and migration. Binding of the actin-linking protein, talin, to integrin β cytoplasmic tails (CTs) induces high affinity ligand binding (integrin activation), whereas binding of another actin-linking protein, filamin, to the integrin β CTs negatively regulates this process by blocking the talin-integrin interaction. Here we show structurally that migfilin, a novel cytoskeletal adaptor highly enriched in the integrin adhesion sites, strongly interacts with the same region in filamin where integrin β CTs bind. We further demonstrate that the migfilin interaction dissociates filamin from integrin and promotes the talin/integrin binding and integrin activation. Migfilin thus acts as a molecular switch to disconnect filamin from integrin for regulating integrin activation and dynamics of extracellular matrix-actin linkage.Cells reside in a protein network, the extracellular matrix (ECM).4 Cell-ECM contact is crucial for many physiological and pathophysiological processes and is primarily mediated by heterodimeric (α/β) transmembrane receptors, the integrins (1). Integrins engage a variety of ECM proteins via their extracellular domains while connecting to the actin cytoskeleton via their small cytoplasmic tails (CTs). The ability of integrins to bind to their ligands is uniquely controlled by the integrin CTs via a process called “inside-out signaling,” i.e. upon cellular stimulation, an integrin, typically expressed in a latent state, can receive intracellular signal(s) at its CT, which transmits through the transmembrane domain to the extracellular domain thereby converting the receptor from a low to a high affinity state (integrin activation). How such long range information transfer is initiated and regulated has been the central topic of integrin/cell adhesion research over the decades (for reviews see Refs. 2-5). Structural/biochemical studies have indicated that the inside-out signaling involves the unclasping of the integrin α/β CT complex (6-9), followed by extensive rearrangement of transmembrane domain and extracellular domain (10-13). Talin, a large actin-linking protein, was found to play a key role in the unclasping process by binding to the integrin β CTs (7-8, 14). Talin activity appears to be controlled by multiple factors or pathways (15-20).Relevant to this study is the role of filamin, another major actin cross-linking protein (21-22), in integrin activation. Filamin was found to share an overlapping binding site on integrin β CTs with talin and thus suppress the talin-integrin interaction (16). Gene silencing of filamin in various cell lines to remove the filamin-integrin connection enhances integrin activation (16, 23), whereas increased filamin-integrin interaction inhibits cell migration (24), a process critically dependent on integrin activation. Together these observations support the notion that filamin binding to integrin serves as a cellular brake to control the dynamics of the integrin activation by inhibiting talin function and ECM-cytoskeleton communication. The mechanism as to how the filamin brake is turned off to promote integrin activation and cell migration is not understood.Filamin is known to contain an N-terminal actin binding domain (ABD) and a long rod-like domain of 24 immunoglobulin-like repeats, of which repeat 21 (IgFLN21) was shown to play a key role in binding to integrin β CTs and blocking the talin-integrin β CT interaction (16). Interestingly, IgFLN21 also recognizes another intracellular protein called migfilin, which has been shown to be an important regulator of integrin-mediated cytoskeletal rearrangements, cell shape change (25), and cell migration (26).In an effort to dissect the complex intermolecular interactions between migfilin, filamin, and integrin, we have undertaken a detailed structural/functional analysis. Using NMR spectroscopy, we have mapped the precise IgFLN21 binding region in migfilin, which is located at the extreme N terminus (residues 1-24) of migfilin (migfilin-N), and we solved solution structure of the IgFLN21-migfilin-N complex. To our surprise, despite little sequence homology, migfilin binds to the same region in IgFLN21 where integrin β CT binds. Detailed NMR and biochemical analyses demonstrate that the migfilin-filamin interaction is an order of magnitude higher than the integrin-filamin interaction and that the migfilin binding to filamin can competitively dissociate filamin from integrin and thus promote the talin-integrin interaction. Using multiple functional approaches, we further show that migfilin, but not its filamin binding defective mutant, significantly enhances integrin activation. These data suggest a novel regulatory pathway in which the binding of filamin to its downstream target migfilin switches off the integrin-filamin connection, thereby promoting talin binding to and activation of integrins.
Keywords:
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