Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Circ Res ; 133(5): 400-411, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37492967

ABSTRACT

BACKGROUND: FLNC (filamin C), a member of the filamin family predominantly expressed in striated muscles, plays a crucial role in bridging the cytoskeleton and ECM (extracellular matrix) in cardiomyocytes, thereby maintaining heart integrity and function. Although genetic variants within the N-terminal ABD (actin-binding domain) of FLNC have been identified in patients with cardiomyopathy, the precise contribution of the actin-binding capability to FLNC's function in mammalian hearts remains poorly understood. METHODS: We conducted in silico analysis of the 3-dimensional structure of mouse FLNC to identify key amino acid residues within the ABD that are essential for FLNC's actin-binding capacity. Subsequently, we performed coimmunoprecipitation and immunofluorescent assays to validate the in silico findings and assess the impact of these mutations on the interactions with other binding partners and the subcellular localization of FLNC. Additionally, we generated and analyzed knock-in mouse models in which the FLNC-actin interaction was completely disrupted by these mutations. RESULTS: Our findings revealed that F93A/L98E mutations completely disrupted FLNC-actin interaction while preserving FLNC's ability to interact with other binding partners ITGB1 (ß1 integrin) and γ-SAG (γ-sarcoglycan), as well as maintaining FLNC subcellular localization. Loss of FLNC-actin interaction in embryonic cardiomyocytes resulted in embryonic lethality and cardiac developmental defects, including ventricular wall malformation and reduced cardiomyocyte proliferation. Moreover, disruption of FLNC-actin interaction in adult cardiomyocytes led to severe dilated cardiomyopathy, enhanced lethality and dysregulation of key cytoskeleton components. CONCLUSIONS: Our data strongly support the crucial role of FLNC as a bridge between actin filaments and ECM through its interactions with actin, ITGB1, γ-SAG, and other associated proteins in cardiomyocytes. Disruption of FLN-actin interaction may result in detachment of actin filaments from the extracellular matrix, ultimately impairing normal cardiac development and function. These findings also provide insights into mechanisms underlying cardiomyopathy associated with genetic variants in FLNC ABD and other regions.


Subject(s)
Actins , Cardiomyopathies , Mice , Animals , Filamins/genetics , Filamins/metabolism , Actins/genetics , Actins/metabolism , Muscle, Skeletal/metabolism , Cardiomyopathies/genetics , Myocytes, Cardiac/metabolism , Mutation , Mammals
2.
Blood ; 141(21): 2629-2641, 2023 05 25.
Article in English | MEDLINE | ID: mdl-36867840

ABSTRACT

The communication of talin-activated integrin αIIbß3 with the cytoskeleton (integrin outside-in signaling) is essential for platelet aggregation, wound healing, and hemostasis. Filamin, a large actin crosslinker and integrin binding partner critical for cell spreading and migration, is implicated as a key regulator of integrin outside-in signaling. However, the current dogma is that filamin, which stabilizes inactive αIIbß3, is displaced from αIIbß3 by talin to promote the integrin activation (inside-out signaling), and how filamin further functions remains unresolved. Here, we show that while associating with the inactive αIIbß3, filamin also associates with the talin-bound active αIIbß3 to mediate platelet spreading. Fluorescence resonance energy transfer-based analysis reveals that while associating with both αIIb and ß3 cytoplasmic tails (CTs) to maintain the inactive αIIbß3, filamin is spatiotemporally rearranged to associate with αIIb CT alone on activated αIIbß3. Consistently, confocal cell imaging indicates that integrin α CT-linked filamin gradually delocalizes from the ß CT-linked focal adhesion marker-vinculin likely because of the separation of integrin α/ß CTs occurring during integrin activation. High-resolution crystal and nuclear magnetic resonance structure determinations unravel that the activated integrin αIIb CT binds to filamin via a striking α-helix→ß-strand transition with a strengthened affinity that is dependent on the integrin-activating membrane environment containing enriched phosphatidylinositol 4,5-bisphosphate. These data suggest a novel integrin αIIb CT-filamin-actin linkage that promotes integrin outside-in signaling. Consistently, disruption of such linkage impairs the activation state of αIIbß3, phosphorylation of focal adhesion kinase/proto-oncogene tyrosine kinase Src, and cell migration. Together, our findings advance the fundamental understanding of integrin outside-in signaling with broad implications in blood physiology and pathology.


Subject(s)
Platelet Glycoprotein GPIIb-IIIa Complex , Platelet Membrane Glycoprotein IIb , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoprotein IIb/metabolism , Actins/metabolism , Filamins/metabolism , Talin/metabolism , Blood Platelets/metabolism
3.
Biomolecules ; 12(2)2022 01 26.
Article in English | MEDLINE | ID: mdl-35204713

ABSTRACT

Plasminogen and its multiple receptors have been implicated in the responses of many different cell types. Among these receptors, histone 2B (H2B) has been shown to play a prominent role in macrophage responses. The contribution of H2B to plasminogen-induced endothelial migration, an event relevant to wound healing and angiogenesis, is unknown. Plasminogen enhanced the migration of endothelial cells, which was inhibited by both Protease-Activated Receptor-1 (PAR1) and 2 (PAR2) antagonists. H2B was detected on viable endothelial cells of venous and arterial origin, and an antibody to H2B that blocks plasminogen binding also inhibited the plasminogen-dependent migration by these cells. The antibody blockade was as effective as PAR1 or PAR2 antagonists in inhibiting endothelial cell migration. In pull-down experiments, H2B formed a complex with both PAR1 and PAR2 but not ß3 integrin, another receptor implicated in endothelial migration in the presence of plasminogen. H2B was found to be associated with clathrin adapator protein, AP2µ (clathrin AP2µ) and ß-arrestin2, which are central to the internationalization/signaling machinery of the PARs. These associations with PAR1-clathrin adaptor AP2µ- and PAR2-ß-arrestin2-dependent internalization/signaling pathways provide a mechanism to link plasminogen to responses such as wound healing and angiogenesis.


Subject(s)
Receptor, PAR-1 , Receptor, PAR-2 , Endothelial Cells/metabolism , Histones/metabolism , Plasminogen/metabolism
4.
J Biol Chem ; 292(20): 8390-8400, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28348077

ABSTRACT

Filamin-mediated linkages between transmembrane receptors (TR) and the actin cytoskeleton are crucial for regulating many cytoskeleton-dependent cellular processes such as cell shape change and migration. A major TR binding site in the immunoglobulin repeat 21 (Ig21) of filamin is masked by the adjacent repeat Ig20, resulting in autoinhibition. The TR binding to this site triggers the relief of Ig20 and protein kinase A (PKA)-mediated phosphorylation of Ser-2152, thereby dynamically regulating the TR-actin linkages. A P2204L mutation in Ig20 reportedly cause frontometaphyseal dysplasia, a skeletal disorder with unknown pathogenesis. We show here that the P2204L mutation impairs a hydrophobic core of Ig20, generating a conformationally fluctuating molten globule-like state. Consequently, unlike in WT filamin, where PKA-mediated Ser-2152 phosphorylation is ligand-dependent, the P2204L mutant is readily accessible to PKA, promoting ligand-independent phosphorylation on Ser-2152. Strong TR peptide ligands from platelet GP1bα and G-protein-coupled receptor MAS effectively bound Ig21 by displacing Ig20 from autoinhibited WT filamin, but surprisingly, the capacity of these ligands to bind the P2204L mutant was much reduced despite the mutation-induced destabilization of the Ig20 structure that supposedly weakens the autoinhibition. Thermodynamic analysis indicated that compared with WT filamin, the conformationally fluctuating state of the Ig20 mutant makes Ig21 enthalpically favorable to bind ligand but with substantial entropic penalty, resulting in total higher free energy and reduced ligand affinity. Overall, our results reveal an unusual structural and thermodynamic basis for the P2204L-induced dysfunction of filamin and frontometaphyseal dysplasia disease.


Subject(s)
Filamins/chemistry , Forehead/abnormalities , Mutation, Missense , Osteochondrodysplasias , Thermodynamics , Amino Acid Substitution , Filamins/genetics , Filamins/metabolism , Humans , Protein Domains
5.
Biochemistry ; 54(44): 6673-83, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26460884

ABSTRACT

Although interaction of a few G protein-coupled receptors (GPCRs) with Filamin A, a key actin cross-linking and biomechanical signal transducer protein, has been observed, a comprehensive structure-function analysis of this interaction is lacking. Through a systematic sequence-based analysis, we found that a conserved filamin binding motif is present in the cytoplasmic domains of >20% of the 824 GPCRs encoded in the human genome. Direct high-affinity interaction of filamin binding motif peptides of select GPCRs with the Ig domain of Filamin A was confirmed by nuclear magnetic resonance spectroscopy and isothermal titration calorimetric experiments. Engagement of the filamin binding motif with the Filamin A Ig domain induced the phosphorylation of filamin by protein kinase A in vitro. In transfected cells, agonist activation as well as constitutive activation of representative GPCRs dramatically elicited recruitment and phosphorylation of cellular Filamin A, a phenomenon long known to be crucial for regulating the structure and dynamics of the cytoskeleton. Our data suggest a molecular mechanism for direct GPCR-cytoskeleton coupling via filamin. Until now, GPCR signaling to the cytoskeleton was predominantly thought to be indirect, through canonical G protein-mediated signaling cascades involving GTPases, adenylyl cyclases, phospholipases, ion channels, and protein kinases. We propose that the GPCR-induced filamin phosphorylation pathway is a conserved, novel biochemical signaling paradigm.


Subject(s)
Filamins/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Binding Sites , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Filamins/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Receptors, G-Protein-Coupled/chemistry , Signal Transduction
6.
Nat Struct Mol Biol ; 22(5): 383-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25849143

ABSTRACT

Activation of heterodimeric (αß) integrin is crucial for regulating cell adhesion. Binding of talin to the cytoplasmic face of integrin activates the receptor, but how integrin is maintained in a resting state to counterbalance its activation has remained obscure. Here, we report the structure of the cytoplasmic domain of human integrin αIIbß3 bound to its inhibitor, the immunoglobin repeat 21 of filamin A (FLNa-Ig21). The structure reveals an unexpected ternary complex in which FLNa-Ig21 not only binds to the C terminus of the integrin ß3 cytoplasmic tail (CT), as previously predicted, but also engages N-terminal helices of αIIb and ß3 CTs to stabilize an inter-CT clasp that helps restrain the integrin in a resting state. Combined with functional data, the structure reveals a new mechanism of filamin-mediated retention of inactive integrin, suggesting a new framework for understanding regulation of integrin activation and adhesion.


Subject(s)
Filamins/metabolism , Filamins/ultrastructure , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/ultrastructure , Cell Adhesion/physiology , Crystallography, X-Ray , Humans , Nuclear Magnetic Resonance, Biomolecular , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Surface Plasmon Resonance , Talin/metabolism
7.
J Biol Chem ; 290(13): 8527-38, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25666618

ABSTRACT

Protein phosphorylation mediates essentially all aspects of cellular life. In humans, this is achieved by ∼500 kinases, each recognizing a specific consensus motif (CM) in the substrates. The majority of CMs are surface-exposed and are thought to be accessible to kinases for phosphorylation. Here we investigated the archetypical protein kinase A (PKA)-mediated phosphorylation of filamin, a major cytoskeletal protein that can adopt an autoinhibited conformation. Surprisingly, autoinhibited filamin is refractory to phosphorylation by PKA on a known Ser(2152) site despite its CM being exposed and the corresponding isolated peptide being readily phosphorylated. Structural analysis revealed that although the CM fits into the PKA active site its surrounding regions sterically clash with the kinase. However, upon ligand binding, filamin undergoes a conformational adjustment, allowing rapid phosphorylation on Ser(2152). These data uncover a novel ligand-induced conformational switch to trigger filamin phosphorylation. They further suggest a substrate shape-dependent filtering mechanism that channels specific exposed CM/kinase recognition in diverse signaling responses.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Filamins/chemistry , Protein Processing, Post-Translational , Amino Acid Sequence , Consensus Sequence , Humans , Molecular Sequence Data , Phosphopeptides/chemistry , Phosphorylation
8.
Protein Sci ; 23(6): 833-42, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24687350

ABSTRACT

Mortalin, a member of the Hsp70-family of molecular chaperones, functions in a variety of processes including mitochondrial protein import and quality control, Fe-S cluster protein biogenesis, mitochondrial homeostasis, and regulation of p53. Mortalin is implicated in regulation of apoptosis, cell stress response, neurodegeneration, and cancer and is a target of the antitumor compound MKT-077. Like other Hsp70-family members, Mortalin consists of a nucleotide-binding domain (NBD) and a substrate-binding domain. We determined the crystal structure of the NBD of human Mortalin at 2.8 Å resolution. Although the Mortalin nucleotide-binding pocket is highly conserved relative to other Hsp70 family members, we find that its nucleotide affinity is weaker than that of Hsc70. A Parkinson's disease-associated mutation is located on the Mortalin-NBD surface and may contribute to Mortalin aggregation. We present structure-based models for how the Mortalin-NBD may interact with the nucleotide exchange factor GrpEL1, with p53, and with MKT-077. Our structure may contribute to the understanding of disease-associated Mortalin mutations and to improved Mortalin-targeting antitumor compounds.


Subject(s)
HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , Mitochondria/metabolism , Humans , Nucleotides/metabolism , Protein Binding
9.
Biochim Biophys Acta ; 1838(2): 579-88, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23891718

ABSTRACT

Cells undergo dynamic remodeling of the cytoskeleton during adhesion and migration on various extracellular matrix (ECM) substrates in response to physiological and pathological cues. The major mediators of such cellular responses are the heterodimeric adhesion receptors, the integrins. Extracellular or intracellular signals emanating from different signaling cascades cause inside-out signaling of integrins via talin, a cystokeletal protein that links integrins to the actin cytoskeleton. Various integrin subfamilies communicate with each other and growth factor receptors under diverse cellular contexts to facilitate or inhibit various integrin-mediated functions. Since talin is an essential mediator of integrin activation, much of the integrin crosstalk would therefore be influenced by talin. However, despite the existence of an extensive body of knowledge on the role of talin in integrin activation and as a stabilizer of ECM-actin linkage, information on its role in regulating inter-integrin communication is limited. This review will focus on the structure of talin, its regulation of integrin activation and discuss its potential role in integrin crosstalk. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.


Subject(s)
Extracellular Matrix/metabolism , Integrins/metabolism , Talin/metabolism , Animals , Humans , Signal Transduction
10.
J Struct Biol ; 179(3): 289-98, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22414427

ABSTRACT

Particularly in higher eukaryotes, some protein domains are found in tandem repeats, performing broad functions often related to cellular organization. For instance, the eukaryotic protein filamin interacts with many proteins and is crucial for the cytoskeleton. The functional properties of long repeat domains are governed by the specific properties of each individual domain as well as by the repeat copy number. To provide better understanding of the evolutionary and functional history of repeating domains, we investigated the mode of evolution of the filamin domain in some detail. Among the domains that are common in long repeat proteins, sushi and spectrin domains evolve primarily through cassette tandem duplications while scavenger and immunoglobulin repeats appear to evolve through clustered tandem duplications. Additionally, immunoglobulin and filamin repeats exhibit a unique pattern where every other domain shows high sequence similarity. This pattern may be the result of tandem duplications, serve to avert aggregation between adjacent domains or it is the result of functional constraints. In filamin, our studies confirm the presence of interspersed integrin binding domains in vertebrates, while invertebrates exhibit more varied patterns, including more clustered integrin binding domains. The most notable case is leech filamin, which contains a 20 repeat expansion and exhibits unique dimerization topology. Clearly, invertebrate filamins are varied and contain examples of similar adjacent integrin-binding domains. Given that invertebrate integrin shows more similarity to the weaker filamin binder, integrin ß3, it is possible that the distance between integrin-binding domains is not as crucial for invertebrate filamins as for vertebrates.


Subject(s)
Contractile Proteins/chemistry , Contractile Proteins/genetics , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Amino Acid Sequence , Animals , Cluster Analysis , Consensus Sequence , Evolution, Molecular , Filamins , Humans , Markov Chains , Models, Genetic , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Repetitive Sequences, Amino Acid , Sequence Analysis, Protein
11.
PLoS One ; 6(10): e26355, 2011.
Article in English | MEDLINE | ID: mdl-22043318

ABSTRACT

Cell adhesion and migration depend on engagement of extracellular matrix ligands by integrins. Integrin activation is dynamically regulated by interactions of various cytoplasmic proteins, such as filamin and integrin activators, talin and kindlin, with the cytoplasmic tail of the integrin ß subunit. Although filamin has been suggested to be an inhibitor of integrin activation, direct functional evidence for the inhibitory role of filamin is limited. Migfilin, a filamin-binding protein enriched at cell-cell and cell-extracellular matrix contact sites, can displace filamin from ß1 and ß3 integrins and promote integrin activation. However, its role in activation and functions of different ß integrins in human vascular cells is unknown. In this study, using flow cytometry, we demonstrate that filamin inhibits ß1 and αIIbß3 integrin activation, and migfilin can overcome its inhibitory effect. Migfilin protein is widely expressed in different adherent and circulating blood cells and can regulate integrin activation in naturally-occurring vascular cells, endothelial cells and neutrophils. Migfilin can activate ß1, ß2 and ß3 integrins and promote integrin mediated responses while migfilin depletion impairs the spreading and migration of endothelial cells. Thus, filamin can act broadly as an inhibitor and migfilin is a promoter of integrin activation.


Subject(s)
Cell Adhesion Molecules/metabolism , Contractile Proteins/metabolism , Cytoskeletal Proteins/metabolism , Endothelial Cells/metabolism , Integrins/metabolism , Microfilament Proteins/metabolism , Neutrophils/metabolism , Cell Adhesion , Endothelium, Vascular , Filamins , Flow Cytometry , Humans , Integrin beta Chains/metabolism
12.
Biochemistry ; 50(20): 4229-31, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21524097

ABSTRACT

Filamin, a large cytoskeletal adaptor, connects plasma membrane to cytoskeleton by binding to transmembrane receptor integrin and actin. Seven of 24 filamin immunoglobulin repeats have conserved integrin binding sites, of which repeats 19 and 21 were shown to be autoinhibited by their adjacent repeats 18 and 20, respectively. Here we show using nuclear magnetic resonance spectroscopy that the autoinhibition can be relieved by integrin or integrin regulator migfilin. We further demonstrate that repeats 19 and 21 can simultaneously engage ligands. The data suggest that filamin is mechanically stretched by integrin or migfilin via a multisite binding mechanism for regulating cytoskeleton and integrin-mediated cell adhesion.


Subject(s)
Contractile Proteins/metabolism , Cytoskeletal Proteins/metabolism , Integrins/metabolism , Microfilament Proteins/metabolism , Calorimetry , Filamins , Humans , Ligands , Nuclear Magnetic Resonance, Biomolecular , Protein Binding
13.
J Biol Chem ; 285(22): 17166-76, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20351101

ABSTRACT

Mutations in the chloride channel cystic fibrosis transmembrane regulator (CFTR) cause cystic fibrosis, a genetic disorder characterized by defects in CFTR biosynthesis, localization to the cell surface, or activation by regulatory factors. It was discovered recently that surface localization of CFTR is stabilized by an interaction between the CFTR N terminus and the multidomain cytoskeletal protein filamin. The details of the CFTR-filamin interaction, however, are unclear. Using x-ray crystallography, we show how the CFTR N terminus binds to immunoglobulin-like repeat 21 of filamin A (FlnA-Ig21). CFTR binds to beta-strands C and D of FlnA-Ig21 using backbone-backbone hydrogen bonds, a linchpin serine residue, and hydrophobic side-chain packing. We use NMR to determine that the CFTR N terminus also binds to several other immunoglobulin-like repeats from filamin A in vitro. Our structural data explain why the cystic fibrosis-causing S13F mutation disrupts CFTR-filamin interaction. We show that FlnA-Ig repeats transfected into cultured Calu-3 cells disrupt CFTR-filamin interaction and reduce surface levels of CFTR. Our findings suggest that filamin A stabilizes surface CFTR by anchoring it to the actin cytoskeleton through interactions with multiple filamin Ig repeats. Such an interaction mode may allow filamins to cluster multiple CFTR molecules and to promote colocalization of CFTR and other filamin-binding proteins in the apical plasma membrane of epithelial cells.


Subject(s)
Contractile Proteins/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Immunoglobulins/chemistry , Microfilament Proteins/chemistry , Actins/chemistry , Amino Acid Sequence , Biotinylation , Cell Membrane/metabolism , Computational Biology/methods , Crystallography, X-Ray/methods , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Filamins , Humans , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Mutation , Protein Structure, Tertiary , Sequence Homology, Amino Acid
14.
J Biol Chem ; 284(49): 34308-20, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19833732

ABSTRACT

Integrin-mediated cell-extracellular matrix (ECM) adhesion is essential for protection of epithelial cells against apoptosis, but the underlying mechanism is incompletely understood. Here we show that migfilin, an integrin-proximal adaptor protein, interacts with Src and contributes to cell-ECM-mediated survival signaling. Loss of cell-ECM adhesion markedly reduces the migfilin level in untransformed epithelial cells and concomitantly induces apoptosis. Overexpression of migfilin substantially desensitizes cell detachment-induced apoptosis. Conversely, depletion of migfilin promotes apoptosis despite the presence of cell-ECM adhesion. At the molecular level migfilin directly interacts with Src, and the migfilin binding surface overlaps with the inhibitory intramolecular interaction sites in Src. Consequently, the binding of migfilin activates Src, resulting in suppression of apoptosis. Our results reveal a novel mechanism by which cell-ECM adhesion regulates Src activation and survival signaling. This migfilin-mediated signaling pathway is dysfunctional in multiple types of carcinoma cells, which likely contributes to aberrant Src activation and anoikis resistance in the cancerous cells.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Gene Expression Regulation , src-Family Kinases/metabolism , Anoikis , Apoptosis , Cell Adhesion , Cell Line, Tumor , Cell Survival , Cytoplasm/metabolism , Epithelial Cells/cytology , Extracellular Matrix/metabolism , Glutathione Transferase/metabolism , Humans , Magnetic Resonance Spectroscopy , RNA Interference , Signal Transduction
15.
J Biol Chem ; 284(50): 35113-21, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19828450

ABSTRACT

The actin-binding protein filamin links membrane receptors to the underlying cytoskeleton. The cytoplasmic domains of these membrane receptors have been shown to bind to various filamin immunoglobulin repeats. Notably, among 24 human filamin repeats, repeat 17 was reported to specifically bind to platelet receptor glycoprotein Ibalpha and repeat 21 to integrins. However, a complete sequence alignment of all 24 human filamin repeats reveals that repeats 17 and 21 actually belong to a distinct filamin repeat subgroup (containing repeats 4, 9, 12, 17, 19, 21, and 23) that shares a conserved ligand-binding site. Using isothermal calorimetry and NMR analyses, we show that all repeats in this subgroup can actually bind glycoprotein Ibalpha, integrins, and a cytoskeleton regulator migfilin in similar manners. These data provide a new view on the ligand specificity of the filamin repeats. They also suggest a multiple ligand binding mechanism where similar repeats within a filamin monomer may promote receptor clustering or receptor cross-talking for regulation of the cytoskeleton organization and diverse filamin-mediated cellular activities.


Subject(s)
Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Binding Sites , Calorimetry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Contractile Proteins/chemistry , Contractile Proteins/classification , Contractile Proteins/genetics , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Filamins , Humans , Integrins/genetics , Integrins/metabolism , Membrane Glycoproteins/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/classification , Microfilament Proteins/genetics , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Phylogeny , Platelet Glycoprotein GPIb-IX Complex , Protein Isoforms/chemistry , Protein Isoforms/classification , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Alignment
16.
J Biol Chem ; 284(7): 4713-22, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19074766

ABSTRACT

The linkage of heterodimeric (alpha/beta) 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 beta cytoplasmic tails (CTs) induces high affinity ligand binding (integrin activation), whereas binding of another actin-linking protein, filamin, to the integrin beta 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 beta 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.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Cytoskeletal Proteins/metabolism , Integrin alpha Chains/metabolism , Integrin beta Chains/metabolism , Actins/genetics , Actins/metabolism , Animals , CHO Cells , Cell Adhesion/physiology , Cell Adhesion Molecules/genetics , Contractile Proteins/genetics , Contractile Proteins/metabolism , Cricetinae , Cricetulus , Cytoskeletal Proteins/genetics , Cytoskeleton/genetics , Cytoskeleton/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Filamins , Humans , Integrin alpha Chains/genetics , Integrin beta Chains/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Talin/genetics , Talin/metabolism
17.
J Magn Reson ; 180(2): 203-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16530439

ABSTRACT

A major step toward the protein structure determination by nuclear magnetic resonance (NMR) spectroscopy is the assignment of multidimensional NMR signals that provide through-bond and through-space inter-atomic correlations. Ambiguities often occur during the assignment process due to resonance degeneracy, which challenges high resolution and larger size protein structure determination. Here, we present a method that will significantly improve the efficiency and accuracy of the NMR signal assignment. The method is based on a correlated accordion principle that, when incorporated into conventional three-dimensional (3D) heteronuclear NMR experiments, allows the retrieval of additional frequency correlation information at high resolution. We show that 3D spectra derived from this method are as effective as the impractical high resolution four-dimensional (4D) spectra with substantially reduced signal ambiguity as compared to their conventional counterparts. The approach promises increased accuracy and size of protein structures determined by NMR.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , Proteins/chemistry , Carbon Isotopes , Nitrogen Isotopes , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...