Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 141(21): 2629-2641, 2023 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-36867840

RESUMO

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.


Assuntos
Complexo Glicoproteico GPIIb-IIIa de Plaquetas , Glicoproteína IIb da Membrana de Plaquetas , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Actinas/metabolismo , Filaminas/metabolismo , Talina/metabolismo , Plaquetas/metabolismo
2.
Circ Res ; 133(5): 400-411, 2023 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-37492967

RESUMO

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.


Assuntos
Actinas , Cardiomiopatias , Camundongos , Animais , Filaminas/genética , Filaminas/metabolismo , Actinas/genética , Actinas/metabolismo , Músculo Esquelético/metabolismo , Cardiomiopatias/genética , Miócitos Cardíacos/metabolismo , Mutação , Mamíferos
3.
J Biol Chem ; 290(13): 8527-38, 2015 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-25666618

RESUMO

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.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/química , Filaminas/química , Processamento de Proteína Pós-Traducional , Sequência de Aminoácidos , Sequência Consenso , Humanos , Dados de Sequência Molecular , Fosfopeptídeos/química , Fosforilação
4.
Biomolecules ; 12(2)2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-35204713

RESUMO

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.


Assuntos
Receptor PAR-1 , Receptor PAR-2 , Células Endoteliais/metabolismo , Histonas/metabolismo , Plasminogênio/metabolismo
5.
Biochemistry ; 50(20): 4229-31, 2011 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-21524097

RESUMO

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.


Assuntos
Proteínas Contráteis/metabolismo , Proteínas do Citoesqueleto/metabolismo , Integrinas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Calorimetria , Filaminas , Humanos , Ligantes , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica
6.
J Biol Chem ; 284(49): 34308-20, 2009 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19833732

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/metabolismo , Membrana Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Regulação da Expressão Gênica , Quinases da Família src/metabolismo , Anoikis , Apoptose , Adesão Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Citoplasma/metabolismo , Células Epiteliais/citologia , Matriz Extracelular/metabolismo , Glutationa Transferase/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Interferência de RNA , Transdução de Sinais
7.
J Biol Chem ; 284(50): 35113-21, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19828450

RESUMO

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.


Assuntos
Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Calorimetria , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Proteínas Contráteis/química , Proteínas Contráteis/classificação , Proteínas Contráteis/genética , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Filaminas , Humanos , Integrinas/genética , Integrinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/classificação , Proteínas dos Microfilamentos/genética , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Filogenia , Complexo Glicoproteico GPIb-IX de Plaquetas , Isoformas de Proteínas/química , Isoformas de Proteínas/classificação , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Alinhamento de Sequência
8.
Nat Struct Mol Biol ; 22(5): 383-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25849143

RESUMO

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.


Assuntos
Filaminas/metabolismo , Filaminas/ultraestrutura , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/ultraestrutura , Adesão Celular/fisiologia , Cristalografia por Raios X , Humanos , Ressonância Magnética Nuclear Biomolecular , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície , Talina/metabolismo
9.
PLoS One ; 6(10): e26355, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22043318

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/metabolismo , Proteínas Contráteis/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células Endoteliais/metabolismo , Integrinas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Neutrófilos/metabolismo , Adesão Celular , Endotélio Vascular , Filaminas , Citometria de Fluxo , Humanos , Cadeias beta de Integrinas/metabolismo
10.
J Biol Chem ; 284(7): 4713-22, 2009 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19074766

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/metabolismo , Movimento Celular/fisiologia , Proteínas do Citoesqueleto/metabolismo , Cadeias alfa de Integrinas/metabolismo , Cadeias beta de Integrinas/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Células CHO , Adesão Celular/fisiologia , Moléculas de Adesão Celular/genética , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Cricetinae , Cricetulus , Proteínas do Citoesqueleto/genética , Citoesqueleto/genética , Citoesqueleto/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Filaminas , Humanos , Cadeias alfa de Integrinas/genética , Cadeias beta de Integrinas/genética , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Talina/genética , Talina/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa