Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Cell Biol ; 25(1): 120-133, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36543981

RESUMO

In response to different types and intensities of mechanical force, cells modulate their physical properties and adapt their plasma membrane (PM). Caveolae are PM nano-invaginations that contribute to mechanoadaptation, buffering tension changes. However, whether core caveolar proteins contribute to PM tension accommodation independently from the caveolar assembly is unknown. Here we provide experimental and computational evidence supporting that caveolin-1 confers deformability and mechanoprotection independently from caveolae, through modulation of PM curvature. Freeze-fracture electron microscopy reveals that caveolin-1 stabilizes non-caveolar invaginations-dolines-capable of responding to low-medium mechanical forces, impacting downstream mechanotransduction and conferring mechanoprotection to cells devoid of caveolae. Upon cavin-1/PTRF binding, doline size is restricted and membrane buffering is limited to relatively high forces, capable of flattening caveolae. Thus, caveolae and dolines constitute two distinct albeit complementary components of a buffering system that allows cells to adapt efficiently to a broad range of mechanical stimuli.


Assuntos
Cavéolas , Caveolina 1 , Cavéolas/metabolismo , Caveolina 1/metabolismo , Mecanotransdução Celular , Membrana Celular/metabolismo , Proteínas/metabolismo
2.
Biomolecules ; 12(3)2022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35327596

RESUMO

Cells have adapted to mechanical forces early in evolution and have developed multiple mechanisms ensuring sensing of, and adaptation to, the diversity of forces operating outside and within organisms. The nucleus must necessarily adapt to all types of mechanical signals, as its functions are essential for virtually all cell processes, many of which are tuned by mechanical cues. To sense forces, the nucleus is physically connected with the cytoskeleton, which senses and transmits forces generated outside and inside the cell. The nuclear LINC complex bridges the cytoskeleton and the nuclear lamina to transmit mechanical information up to the chromatin. This system creates a force-sensing macromolecular complex that, however, is not sufficient to regulate all nuclear mechanoadaptation processes. Within the nucleus, additional mechanosensitive structures, including the nuclear envelope and the nuclear pore complex, function to regulate nuclear mechanoadaptation. Similarly, extra nuclear mechanosensitive systems based on plasma membrane dynamics, mechanotransduce information to the nucleus. Thus, the nucleus has the intrinsic structural components needed to receive and interpret mechanical inputs, but also rely on extra nuclear mechano-sensors that activate nuclear regulators in response to force. Thus, a network of mechanosensitive cell structures ensures that the nucleus has a tunable response to mechanical cues.


Assuntos
Núcleo Celular , Membrana Nuclear , Núcleo Celular/metabolismo , Cromatina/metabolismo , Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Membrana Nuclear/metabolismo
3.
Nat Commun ; 13(1): 1174, 2022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35246520

RESUMO

Mechanical forces regulate multiple essential pathways in the cell. The nuclear translocation of mechanoresponsive transcriptional regulators is an essential step for mechanotransduction. However, how mechanical forces regulate the nuclear import process is not understood. Here, we identify a highly mechanoresponsive nuclear transport receptor (NTR), Importin-7 (Imp7), that drives the nuclear import of YAP, a key regulator of mechanotransduction pathways. Unexpectedly, YAP governs the mechanoresponse of Imp7 by forming a YAP/Imp7 complex that responds to mechanical cues through the Hippo kinases MST1/2. Furthermore, YAP behaves as a dominant cargo of Imp7, restricting the Imp7 binding and the nuclear translocation of other Imp7 cargoes such as Smad3 and Erk2. Thus, the nuclear import process is an additional regulatory layer indirectly regulated by mechanical cues, which activate a preferential Imp7 cargo, YAP, which competes out other cargoes, resulting in signaling crosstalk.


Assuntos
Núcleo Celular , Mecanotransdução Celular , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Carioferinas/genética , Carioferinas/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo
4.
Curr Opin Cell Biol ; 68: 113-123, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33188985

RESUMO

Mechanical forces (extracellular matrix stiffness, vascular shear stress, and muscle stretching) reaching the plasma membrane (PM) determine cell behavior. Caveolae are PM-invaginated nanodomains with specific lipid and protein composition. Being highly abundant in mechanically challenged tissues (muscles, lungs, vessels, and adipose tissues), they protect cells from mechanical stress damage. Caveolae flatten upon increased PM tension, enabling both force sensing and accommodation, critical for cell mechanoprotection and homeostasis. Thus, caveolae are highly plastic, ranging in complexity from flattened membranes to vacuolar invaginations surrounded by caveolae-rosettes-which also contribute to mechanoprotection. Caveolar components crosstalk with mechanotransduction pathways and recent studies show that they translocate from the PM to the nucleus to convey stress information. Furthermore, caveolae components can regulate membrane traffic from/to the PM to adapt to environmental mechanical forces. The interdependence between lipids and caveolae starts to be understood, and the relevance of caveolae-dependent membrane trafficking linked to mechanoadaption to different physiopathological processes is emerging.


Assuntos
Transporte Biológico , Cavéolas/metabolismo , Membrana Celular/metabolismo , Mecanotransdução Celular , Animais , Endocitose , Matriz Extracelular/metabolismo , Humanos , Estresse Mecânico
5.
Nat Commun ; 10(1): 5828, 2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31862885

RESUMO

Cells remodel their structure in response to mechanical strain. However, how mechanical forces are translated into biochemical signals that coordinate the structural changes observed at the plasma membrane (PM) and the underlying cytoskeleton during mechanoadaptation is unclear. Here, we show that PM mechanoadaptation is controlled by a tension-sensing pathway composed of c-Abl tyrosine kinase and membrane curvature regulator FBP17. FBP17 is recruited to caveolae to induce the formation of caveolar rosettes. FBP17 deficient cells have reduced rosette density, lack PM tension buffering capacity under osmotic shock, and cannot adapt to mechanical strain. Mechanistically, tension is transduced to the FBP17 F-BAR domain by direct phosphorylation mediated by c-Abl, a mechanosensitive molecule. This modification inhibits FBP17 membrane bending activity and releases FBP17-controlled inhibition of mDia1-dependent stress fibers, favoring membrane adaptation to increased tension. This mechanoprotective mechanism adapts the cell to changes in mechanical tension by coupling PM and actin cytoskeleton remodeling.


Assuntos
Cavéolas/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo , Mecanotransdução Celular , Proteínas Proto-Oncogênicas c-abl/metabolismo , Fibras de Estresse/metabolismo , Cavéolas/ultraestrutura , Proteínas de Ligação a Ácido Graxo/genética , Fibroblastos , Técnicas de Inativação de Genes , Células HEK293 , Células HeLa , Humanos , Microscopia Eletrônica , Fosforilação , RNA Interferente Pequeno/metabolismo , Fibras de Estresse/ultraestrutura , Estresse Mecânico
6.
Methods Mol Biol ; 1487: 163-174, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27924566

RESUMO

Caveolin1, the main component of caveolae, plays a major role in regulating cell motility, gene expression, and cytoskeleton remodeling downstream of many membrane receptors. Here, we summarize different techniques set up to study changes in cell morphology and cell motility regulated by ERK/caveolin1 interactions during induction of epithelial mesenchymal transition (EMT) in mesothelial cells (MCs).


Assuntos
Caveolina 1/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Transdução de Sinais , Animais , Técnicas de Cultura de Células , Movimento Celular , Forma Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal , Camundongos , Cavidade Peritoneal , Ligação Proteica
7.
J Cell Sci ; 128(15): 2747-58, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26159735

RESUMO

An essential property of the plasma membrane of mammalian cells is its plasticity, which is required for sensing and transmitting of signals, and for accommodating the tensional changes imposed by its environment or its own biomechanics. Caveolae are unique invaginated membrane nanodomains that play a major role in organizing signaling, lipid homeostasis and adaptation to membrane tension. Caveolae are frequently associated with stress fibers, a major regulator of membrane tension and cell shape. In this Commentary, we discuss recent studies that have provided new insights into the function of caveolae and have shown that trafficking and organization of caveolae are tightly regulated by stress-fiber regulators, providing a functional link between caveolae and stress fibers. Furthermore, the tension in the plasma membrane determines the curvature of caveolae because they flatten at high tension and invaginate at low tension, thus providing a tension-buffering system. Caveolae also regulate multiple cellular pathways, including RhoA-driven actomyosin contractility and other mechanosensitive pathways, suggesting that caveolae could couple mechanotransduction pathways to actin-controlled changes in tension through their association with stress fibers. Therefore, we argue here that the association of caveolae with stress fibers could provide an important strategy for cells to deal with mechanical stress.


Assuntos
Fenômenos Biomecânicos/fisiologia , Cavéolas/metabolismo , Mecanotransdução Celular/fisiologia , Fibras de Estresse/metabolismo , Estresse Mecânico , Actomiosina/metabolismo , Animais , Membrana Celular/fisiologia , Humanos , Estrutura Terciária de Proteína , Transporte Proteico , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo
8.
Cell Commun Signal ; 12: 57, 2014 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-25238970

RESUMO

BACKGROUND: Adhesion and migration are relevant physiological functions that must be regulated by the cell under both normal and pathological conditions. The dioxin receptor (AhR) has emerged as a transcription factor regulating both processes in mesenchymal, epithelial and endothelial cells. Indirect results suggest that AhR could cooperate not only with additional transcription factors but also with membrane-associated proteins to drive such processes. RESULTS: In this study, we have used immortalized and primary dermal fibroblasts from wild type (AhR+/+) and AhR-null (AhR-/-) mice to show that AhR modulates membrane distribution and mobilization of caveolin-1 (Cav-1) during directional cell migration. AhR co-immunoprecipitated with Cav-1 and a fraction of both proteins co-localized to detergent-resistant membrane microdomains (DRM). Consistent with a role of AhR in the process, AhR-/- cells had a significant reduction in Cav-1 in DRMs. Moreover, high cell density reduced AhR nuclear levels and moved Cav-1 from DRMs to the soluble membrane in AhR+/+ but not in AhR-/- cells. Tyrosine-14 phosphorylation had a complex role in the mechanism since its upregulation reduced Cav-1 in DRMs in both AhR+/+ and AhR-/-cells, despite the lower basal levels of Y14-Cav-1 in the null cells. Fluorescence recovery after photobleaching revealed that AhR knock-down blocked Cav-1 transport to the plasma membrane, a deficit possibly influencing its depleted levels in DRMs. Membrane distribution of Cav-1 in AhR-null fibroblasts correlated with higher levels of cholesterol and with disrupted membrane microdomains, whereas addition of exogenous cholesterol changed the Cav-1 distribution of AhR+/+ cells to the null phenotype. Consistently, higher cholesterol levels enhanced caveolae-dependent endocytosis in AhR-null cells. CONCLUSIONS: These results suggest that AhR modulates Cav-1 distribution in migrating cells through the control of cholesterol-enriched membrane microdomains. Our study also supports the likely possibility of membrane-related, transcription factor independent, functions of AhR.


Assuntos
Caveolina 1/metabolismo , Movimento Celular/fisiologia , Colesterol/metabolismo , Fibroblastos/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Células Cultivadas , Endocitose , Fibroblastos/fisiologia , Camundongos , Camundongos Knockout , Receptores de Hidrocarboneto Arílico/genética
9.
J Cell Sci ; 125(Pt 13): 3097-113, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22454521

RESUMO

The biology of caveolin-1 (Cav1)/caveolae is intimately linked to actin dynamics and adhesion receptors. Caveolar domains are organized in hierarchical levels of complexity from curved or flattened caveolae to large, higher-order caveolar rosettes. We report that stress fibers controlled by Abl kinases and mDia1 determine the level of caveolar domain organization, which conditions the subsequent inward trafficking of caveolar domains induced upon loss of cell adhesion from the extracellular matrix. Abl-deficient cells have fewer stress fibers, a smaller pool of stress-fiber co-aligned Cav1 and increased clustering of Cav1/caveolae at the cell surface. Defective caveolar linkage to stress fibers prevents the formation of big caveolar rosettes upon loss of cell adhesion, correlating with a lack of inward trafficking. Live imaging of stress fibers and Cav1 showed that the actin-linked Cav1 pool loses its spatial organization in the absence of actin polymerization and is dragged and clustered by depolymerizing filaments. We identified mDia1 as the actin polymerization regulator downstream of Abl kinases that controls the stress-fiber-linked Cav1 pool. mDia1 knockdown results in Cav1/caveolae clustering and defective inward trafficking upon loss of cell adhesion. By contrast, cell elongation imposed by the excess of stress fibers induced by active mDia1 flattens caveolae. Furthermore, active mDia1 rescues the actin co-aligned Cav1 pool and Cav1 inward trafficking upon loss of adhesion in Abl-deficient cells. Thus, caveolar domain organization and trafficking are tightly coupled to adhesive and stress fiber regulatory pathways.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cavéolas/metabolismo , Caveolina 1/metabolismo , Proteínas Tirosina Quinases/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Cavéolas/fisiologia , Cavéolas/ultraestrutura , Caveolina 1/genética , Adesão Celular , Clonagem Molecular , Forminas , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos , Microscopia Eletrônica , Plasmídeos/genética , Plasmídeos/metabolismo , Polimerização , Estrutura Terciária de Proteína , Transporte Proteico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fibras de Estresse/metabolismo , Fibras de Estresse/fisiologia
11.
J Cell Sci ; 124(Pt 16): 2763-76, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21807941

RESUMO

Caveolae are relatively stable membrane invaginations that compartmentalize signaling, regulate lipid metabolism and mediate viral entry. Caveolae are closely associated with actin fibers and internalize in response to diverse stimuli. Loss of cell adhesion is known to induce rapid and robust caveolae internalization and trafficking toward a Rab11-positive recycling endosome; however, pathways governing this process are poorly understood. Here, we report that filamin A is required to maintain the F-actin-dependent linear distribution of caveolin-1. High spatiotemporal resolution particle tracking of caveolin-1-GFP vesicles by total internal reflection fluorescence (TIRF) microscopy revealed that FLNa is required for the F-actin-dependent arrest of caveolin-1 vesicles in a confined area and their stable anchorage to the plasma membrane. The linear distribution and anchorage of caveolin-1 vesicles are both required for proper caveolin-1 inwards trafficking. De-adhesion-triggered caveolae inward trafficking towards a recycling endosome is impaired in FLNa-depleted HeLa and FLNa-deficient M2-melanoma cells. Inwards trafficking of caveolin-1 requires both the ability of FLNa to bind actin and cycling PKCα-dependent phosphorylation of FLNa on Ser2152 after cell detachment.


Assuntos
Actinas/metabolismo , Cavéolas/metabolismo , Caveolina 1/metabolismo , Membrana Celular/metabolismo , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Cavéolas/ultraestrutura , Adesão Celular , Proteínas Contráteis/genética , Endossomos/metabolismo , Filaminas , Células HeLa , Humanos , Proteínas dos Microfilamentos/genética , Microscopia de Interferência , Fosforilação/genética , Ligação Proteica/genética , Proteína Quinase C/metabolismo , Transporte Proteico , RNA Interferente Pequeno/genética
12.
Cell ; 146(1): 148-63, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21729786

RESUMO

Mechanotransduction is a key determinant of tissue homeostasis and tumor progression. It is driven by intercellular adhesions, cell contractility, and forces generated within the microenvironment and is dependent on extracellular matrix composition, organization, and compliance. We show that caveolin-1 (Cav1) favors cell elongation in three-dimensional cultures and promotes Rho- and force-dependent contraction, matrix alignment, and microenvironment stiffening through regulation of p190RhoGAP. In turn, microenvironment remodeling by Cav1 fibroblasts forces cell elongation. Cav1-deficient mice have disorganized stromal tissue architecture. Stroma associated with human carcinomas and melanoma metastases is enriched in Cav1-expressing carcinoma-associated fibroblasts (CAFs). Cav1 expression in breast CAFs correlates with low survival, and Cav1 depletion in CAFs decreases CAF contractility. Consistently, fibroblast expression of Cav1, through p190RhoGAP regulation, favors directional migration and invasiveness of carcinoma cells in vitro. In vivo, stromal Cav1 remodels peri- and intratumoral microenvironments to facilitate tumor invasion, correlating with increased metastatic potency. Thus, Cav1 modulates tissue responses through force-dependent architectural regulation of the microenvironment.


Assuntos
Caveolina 1/metabolismo , Metástase Neoplásica/patologia , Neoplasias/patologia , Animais , Movimento Celular , Fibroblastos/patologia , Humanos , Melanoma/patologia , Camundongos , Camundongos Knockout
13.
Mol Cell Biol ; 29(7): 1735-48, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19158278

RESUMO

Actin polymerization provides the driving force for the formation of cell-cell junctions and is mediated by two types of actin nucleators, Arp2/3 and formins. Proteins required for coordinately linking cadherin-mediated adhesion to Arp2/3-dependent versus formin-dependent nucleation have yet to be defined. Here we show a role for Abi, the Abi-binding partner Nap1, and the Nap1-binding protein Sra1 in the regulation of cadherin-dependent adhesion. We found that Abi, which is known to interact with Wave, leading to activation of the Arp2/3 complex, is also capable of interacting with the Diaphanous (Dia)-related formins in the absence of Wave. Knockdown of Abi, Nap1, Sra1, or Dia markedly inhibited cell-cell junctions, whereas knockdown of Wave or Arp2/3 produced mild and transient phenotypes. Dia and Abi colocalized with beta-catenin at cell-cell junctions. Further, Dia and Wave bound to overlapping sites on Abi1, and Wave competed with Dia for Abi1 binding. Notably, an active Dia1 C-terminal fragment that localizes to cell-cell junctions rescued the abnormal junctions induced by depletion of Abi or Nap1 in epithelial cells. These findings uncover a novel link between cadherin-mediated adhesion and the regulation of actin dynamics through the requirement for an Abi/Dia complex for the formation and stability of cell-cell junctions.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas do Citoesqueleto/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Animais , Caderinas/metabolismo , Proteínas de Transporte/metabolismo , Adesão Celular , Linhagem Celular , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/deficiência , Cães , Proteínas Fetais/metabolismo , Forminas , Humanos , Junções Intercelulares/metabolismo , Camundongos , Proteínas dos Microfilamentos/metabolismo , Proteínas Nucleares/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas/metabolismo , Fatores de Tempo , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , tRNA Metiltransferases
14.
Semin Cell Dev Biol ; 18(5): 627-37, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17904396

RESUMO

Cells have a complex system for delivering and compartmentalizing proteins and lipids in order to achieve spatio-temporal coordination of signaling. Rafts/caveolae are plasma membrane microdomains that regulate signaling pathways and processes such as cell migration, polarization and proliferation. Regulation of raft/caveolae trafficking involves multiple steps regulated by different proteins to ensure coordination of signaling cascades. The best studied raft-mediated endocytic route is controlled by caveolins. Recent data suggest integrin-mediated cell adhesion is a key regulator of caveolar endocytosis. In this review we examine the regulation of caveolar trafficking and the interplay between integrins, cell adhesion and caveolae internalization.


Assuntos
Cavéolas/fisiologia , Exocitose/fisiologia , Integrinas/fisiologia , Microdomínios da Membrana/fisiologia , Transdução de Sinais/fisiologia , Animais , Transporte Biológico/fisiologia , Adesão Celular/fisiologia , Citoesqueleto/fisiologia , Humanos , Modelos Imunológicos
15.
J Cell Biol ; 177(4): 683-94, 2007 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-17517963

RESUMO

Development, angiogenesis, wound healing, and metastasis all involve the movement of cells in response to changes in the extracellular environment. To determine whether caveolin-1 plays a role in cell migration, we have used fibroblasts from knockout mice. Caveolin-1-deficient cells lose normal cell polarity, exhibit impaired wound healing, and have decreased Rho and increased Rac and Cdc42 GTPase activities. Directional persistency of migration is lost, and the cells show an impaired response to external directional stimuli. Both Src inactivation and p190RhoGAP knockdown restore the wild-type phenotype to caveolin-1-deficient cells, suggesting that caveolin-1 stimulates normal Rho GTP loading through inactivation of the Src-p190RhoGAP pathway. These findings highlight the importance of caveolin-1 in the establishment of cell polarity during directional migration through coordination of the signaling of Src kinase and Rho GTPases.


Assuntos
Caveolina 1/fisiologia , Movimento Celular/fisiologia , Polaridade Celular/fisiologia , Proteínas rho de Ligação ao GTP/fisiologia , Quinases da Família src/fisiologia , Animais , Caveolina 1/deficiência , Caveolina 1/genética , Linhagem Celular , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/enzimologia , Fibroblastos/fisiologia , Humanos , Camundongos , Camundongos Knockout , Células NIH 3T3 , Transdução de Sinais/fisiologia
16.
Cell Cycle ; 5(19): 2179-82, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16969102

RESUMO

Integrin-mediated adhesion regulates trafficking of cholesterol-enriched membrane microdomains (CEMM). Upon cell detachment from the extracellular matrix (ECM), CEMMs undergo rapid internalization and are cleared from the plasma membrane. This pathway regulates integrin-mediated Rac membrane targeting, allowing coupling of Rac to downstream effectors. Internalization of CEMMs is mediated by Dynamin-2, a regulator of caveolae dynamics, and caveolin-1, an essential caveolae coat protein. Translocation of tyrosine phosphorylated caveolin-1 from focal adhesions to caveolae upon cell detachment induces CEMM internalization. Notably, integrin-mediated regulation of Erk, phosphatidylinositol-3-OH kinase (PI3K) and Rac pathways is dependent on caveolin-1. These results describe a novel pathway in which integrins prevent downregulation of Erk, PI3K and Rac-dependent pathways by inhibiting caveolin-1-dependent endocytosis. This pathway define a novel molecular mechanism for regulated cell growth and tumor suppression by caveolin-1.


Assuntos
Cavéolas/fisiologia , Endocitose/fisiologia , Integrinas/metabolismo , Transdução de Sinais , Animais , Cavéolas/metabolismo , Humanos
17.
Mol Cell Biol ; 24(24): 10905-22, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15572692

RESUMO

The Abl-interactor (Abi) family of adaptor proteins has been linked to signaling pathways involving the Abl tyrosine kinases and the Rac GTPase. Abi proteins localize to sites of actin polymerization in protrusive membrane structures and regulate actin dynamics in vitro. Here we demonstrate that Abi2 modulates cell morphogenesis and migration in vivo. Homozygous deletion of murine abi2 produced abnormal phenotypes in the eye and brain, the tissues with the highest Abi2 expression. In the absence of Abi2, secondary lens fiber orientation and migration were defective in the eye, without detectable defects in proliferation, differentiation, or apoptosis. These phenotypes were consistent with the localization of Abi2 at adherens junctions in the developing lens and at nascent epithelial cell adherens junctions in vitro. Downregulation of Abi expression by RNA interference impaired adherens junction formation and correlated with downregulation of the Wave actin-nucleation promoting factor. Loss of Abi2 also resulted in cell migration defects in the neocortex and hippocampus, abnormal dendritic spine morphology and density, and severe deficits in short- and long-term memory. These findings support a role for Abi2 in the regulation of cytoskeletal dynamics at adherens junctions and dendritic spines, which is critical for intercellular connectivity, cell morphogenesis, and cognitive functions.


Assuntos
Movimento Celular/genética , Espinhas Dendríticas/genética , Proteínas de Homeodomínio/genética , Aprendizagem , Memória , Morfogênese/genética , Proteínas Adaptadoras de Transdução de Sinal , Junções Aderentes/metabolismo , Animais , Linhagem Celular , Dendritos/genética , Cães , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Deleção de Genes , Células HeLa , Hipocampo/citologia , Proteínas de Homeodomínio/metabolismo , Homozigoto , Humanos , Cristalino/embriologia , Cristalino/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neocórtex/citologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo
18.
Mol Cell Biol ; 24(11): 4979-93, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15143189

RESUMO

The Abl interactor 1 (Abi-1) protein has been implicated in the regulation of actin dynamics and localizes to the tips of lamellipodia and filopodia. Here, we show that Abi-1 binds the actin nucleator protein Wave-1 through an amino-terminal Wave-binding (WAB) domain and that disruption of the Abi-1-Wave-1 interaction prevents Abi-1 from reaching the tip of the lamellipodium. Abi-1 binds to the Wave homology domain of Wave-1, a region that is required for translocation of Wave-1 to the lamellipodium. Mouse embryo fibroblasts that lack one allele of Abi-1 and are homozygous null for the related Abi-2 protein exhibit decreased Wave-1 protein levels. This phenotype is rescued by Abi-1 proteins that retain Wave-1 binding but not by Abi-1 mutants that cannot bind to Wave-1. Moreover, we uncovered an overlapping SNARE domain in the amino terminus of Abi-1 that interacts with Syntaxin-1, a SNARE family member. Further, we demonstrated that Abi-1 shuttles in and out of the nucleus in a leptomycin B (LMB)-dependent manner and that complete nuclear translocation of Abi-1 in the absence of LMB requires the combined inactivation of the SNARE, WAB, and SH3 domains of Abi-1. Thus, Abi-1 undergoes nucleocytoplasmic shuttling and functions at the leading edge to regulate Wave-1 localization and protein levels.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Antígenos de Superfície/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Transporte Vesicular , Sequência de Aminoácidos , Animais , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Estrutura Terciária de Proteína , Proteínas SNARE , Sintaxina 1 , Família de Proteínas da Síndrome de Wiskott-Aldrich
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...