Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38712197

RESUMO

Spatial and temporal protein tracking in live cells permits proteome remodeling in response to extracellular cues. Historically, protein dynamics during trafficking have been visualized using constitutively active fluorescent proteins (FPs) fused to proteins of interest. While powerful, such FPs label all cellular pools of a protein, potentially masking the dynamics of select subpopulations. To help study protein subpopulations, bioconjugate tags, including the fluorogen activation proteins (FAPs), were developed. FAPs are comprised of two components: a single chain antibody (SCA) fused to the protein of interest and a malachite-green (MG) derivative, which fluoresces only when bound to the SCA. Importantly, the MG derivatives can be either cell-permeant or -impermeant, thus permitting isolated detection of SCA-tagged proteins at the cell surface and facilitating quantitative endocytic measures. To expand FAP use in yeast, we optimized the SCA for yeast expression, created FAP-tagging plasmids, and generated FAP-tagged organelle markers. To demonstrate FAP efficacy, we coupled the SCA to the yeast G-protein coupled receptor Ste3. We measured Ste3 endocytic dynamics in response to pheromone and characterized cis- and trans-acting regulators of Ste3. Our work significantly expands FAP technology for varied applications in S. cerevisiae.

2.
J Biol Chem ; 296: 100582, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33771561

RESUMO

α-Catenin binds directly to ß-catenin and connects the cadherin-catenin complex to the actin cytoskeleton. Tension regulates α-catenin conformation. Actomyosin-generated force stretches the middle (M)-region to relieve autoinhibition and reveal a binding site for the actin-binding protein vinculin. It is not known whether the intramolecular interactions that regulate epithelial (αE)-catenin binding are conserved across the α-catenin family. Here, we describe the biochemical properties of testes (αT)-catenin, an α-catenin isoform critical for cardiac function and how intramolecular interactions regulate vinculin-binding autoinhibition. Isothermal titration calorimetry showed that αT-catenin binds the ß-catenin-N-cadherin complex with a similar low nanomolar affinity to that of αE-catenin. Limited proteolysis revealed that the αT-catenin M-region adopts a more open conformation than αE-catenin. The αT-catenin M-region binds the vinculin N-terminus with low nanomolar affinity, indicating that the isolated αT-catenin M-region is not autoinhibited and thereby distinct from αE-catenin. However, the αT-catenin head (N- and M-regions) binds vinculin 1000-fold more weakly (low micromolar affinity), indicating that the N-terminus regulates the M-region binding to vinculin. In cells, αT-catenin recruitment of vinculin to cell-cell contacts requires the actin-binding domain and actomyosin-generated tension, indicating that force regulates vinculin binding. Together, our results show that the αT-catenin N-terminus is required to maintain M-region autoinhibition and modulate vinculin binding. We postulate that the unique molecular properties of αT-catenin allow it to function as a scaffold for building specific adhesion complexes.


Assuntos
Vinculina/metabolismo , alfa Catenina/metabolismo , Citoesqueleto de Actina/metabolismo , Sítios de Ligação , Miocárdio/metabolismo , Ligação Proteica , Proteólise , alfa Catenina/química
3.
Mol Biol Cell ; 30(21): 2639-2650, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31483697

RESUMO

The adherens junction (AJ) couples the actin cytoskeletons of neighboring cells to allow mechanical integration and tissue organization. The physiological demands of intercellular adhesion require that the AJ be responsive to dynamic changes in force while maintaining mechanical load. These demands are tested in the heart, where cardiomyocyte AJs must withstand repeated cycles of actomyosin-mediated contractile force. Here we show that force-responsive cardiomyocyte AJs recruit actin-binding ligands to selectively couple actin networks. We employed a panel of N-cadherin-αE-catenin fusion proteins to rebuild AJs with specific actin linkages in N-cadherin-null cardiomyocytes. In this system, vinculin recruitment was required to rescue myofibril integration at nascent contacts. In contrast, loss of vinculin from the AJ disrupted junction morphology and blocked myofibril integration at cell-cell contacts. Our results identify vinculin as a critical link to contractile actomyosin and offer insight to how actin integration at the AJ is regulated to provide stability under mechanical load.


Assuntos
Citoesqueleto de Actina/metabolismo , Junções Aderentes/metabolismo , Miócitos Cardíacos/metabolismo , Vinculina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Animais , Animais Recém-Nascidos , Caderinas/genética , Caderinas/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Camundongos , Miócitos Cardíacos/citologia , Ligação Proteica , Vinculina/genética , alfa Catenina/genética , alfa Catenina/metabolismo
4.
Mol Biol Cell ; 30(16): 1930-1937, 2019 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-31318313

RESUMO

αT (Testes)-catenin, a critical factor regulating cell-cell adhesion in the heart, directly couples the cadherin-catenin complex to the actin cytoskeleton at the intercalated disk (ICD), a unique cell-cell junction that couples cardiomyocytes. Loss of αT-catenin in mice reduces plakophilin2 and connexin 43 recruitment to the ICD. Since αT-catenin is subjected to mechanical stretch during actomyosin contraction in cardiomyocytes, its activity could be regulated by mechanical force. To provide insight in how force regulates αT-catenin function, we investigated the mechanical stability of the putative, force-sensing middle (M) domain of αT-catenin and determined how force impacts vinculin binding to αT-catenin. We show that 1) physiological levels of force, <15 pN, are sufficient to unfold the three M domains; 2) the M1 domain that harbors the vinculin-binding site is unfolded at ∼6 pN; and 3) unfolding of the M1 domain is necessary for high-affinity vinculin binding. In addition, we quantified the binding kinetics and affinity of vinculin to the mechanically exposed binding site in M1 and observed that αT-catenin binds vinculin with low nanomolar affinity. These results provide important new insights into the mechanosensing properties of αT-catenin and how αT-catenin regulates cell-cell adhesion at the cardiomyocyte ICD.


Assuntos
Vinculina/metabolismo , alfa Catenina/metabolismo , Fenômenos Biomecânicos , Ligação Proteica , Domínios Proteicos , Estabilidade Proteica , alfa Catenina/química
5.
PLoS Genet ; 15(2): e1007720, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30763317

RESUMO

The adherens junction couples the actin cytoskeletons of neighboring cells to provide the foundation for multicellular organization. The core of the adherens junction is the cadherin-catenin complex that arose early in the evolution of multicellularity to link actin to intercellular adhesions. Over time, evolutionary pressures have shaped the signaling and mechanical functions of the adherens junction to meet specific developmental and physiological demands. Evolutionary rate covariation (ERC) identifies proteins with correlated fluctuations in evolutionary rate that can reflect shared selective pressures and functions. Here we use ERC to identify proteins with evolutionary histories similar to the Drosophila E-cadherin (DE-cad) ortholog. Core adherens junction components α-catenin and p120-catenin displayed positive ERC correlations with DE-cad, indicating that they evolved under similar selective pressures during evolution between Drosophila species. Further analysis of the DE-cad ERC profile revealed a collection of proteins not previously associated with DE-cad function or cadherin-mediated adhesion. We then analyzed the function of a subset of ERC-identified candidates by RNAi during border cell (BC) migration and identified novel genes that function to regulate DE-cad. Among these, we found that the gene CG42684, which encodes a putative GTPase activating protein (GAP), regulates BC migration and adhesion. We named CG42684 raskol ("to split" in Russian) and show that it regulates DE-cad levels and actin protrusions in BCs. We propose that Raskol functions with DE-cad to restrict Ras/Rho signaling and help guide BC migration. Our results demonstrate that a coordinated selective pressure has shaped the adherens junction and this can be leveraged to identify novel components of the complexes and signaling pathways that regulate cadherin-mediated adhesion.


Assuntos
Actinas/metabolismo , Caderinas/metabolismo , Adesão Celular/fisiologia , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Citoesqueleto de Actina/metabolismo , Junções Aderentes/metabolismo , Animais , Membrana Celular/metabolismo , Movimento Celular/fisiologia , Transdução de Sinais/fisiologia
6.
J Cell Sci ; 132(3)2019 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-30630894

RESUMO

The junctional complexes that couple cardiomyocytes must transmit the mechanical forces of contraction while maintaining adhesive homeostasis. The adherens junction (AJ) connects the actomyosin networks of neighboring cardiomyocytes and is required for proper heart function. Yet little is known about the molecular composition of the cardiomyocyte AJ or how it is organized to function under mechanical load. Here, we define the architecture, dynamics and proteome of the cardiomyocyte AJ. Mouse neonatal cardiomyocytes assemble stable AJs along intercellular contacts with organizational and structural hallmarks similar to mature contacts. We combine quantitative mass spectrometry with proximity labeling to identify the N-cadherin (CDH2) interactome. We define over 350 proteins in this interactome, nearly 200 of which are unique to CDH2 and not part of the E-cadherin (CDH1) interactome. CDH2-specific interactors comprise primarily adaptor and adhesion proteins that promote junction specialization. Our results provide novel insight into the cardiomyocyte AJ and offer a proteomic atlas for defining the molecular complexes that regulate cardiomyocyte intercellular adhesion. This article has an associated First Person interview with the first authors of the paper.


Assuntos
Citoesqueleto de Actina/metabolismo , Actomiosina/genética , Junções Aderentes/metabolismo , Caderinas/genética , Mecanotransdução Celular , Miócitos Cardíacos/metabolismo , Citoesqueleto de Actina/ultraestrutura , Actomiosina/metabolismo , Junções Aderentes/ultraestrutura , Animais , Animais Recém-Nascidos , Caderinas/metabolismo , Adesão Celular , Comunicação Celular , Regulação da Expressão Gênica , Ontologia Genética , Camundongos , Anotação de Sequência Molecular , Miócitos Cardíacos/ultraestrutura , Cultura Primária de Células , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteômica/métodos
7.
J Biol Chem ; 293(28): 11006-11021, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-29784874

RESUMO

Protein composition at the plasma membrane is tightly regulated, with rapid protein internalization and selective targeting to the cell surface occurring in response to environmental changes. For example, ion channels are dynamically relocalized to or from the plasma membrane in response to physiological alterations, allowing cells and organisms to maintain osmotic and salt homeostasis. To identify additional factors that regulate the selective trafficking of a specific ion channel, we used a yeast model for a mammalian potassium channel, the K+ inward rectifying channel Kir2.1. Kir2.1 maintains potassium homeostasis in heart muscle cells, and Kir2.1 defects lead to human disease. By examining the ability of Kir2.1 to rescue the growth of yeast cells lacking endogenous potassium channels, we discovered that specific α-arrestins regulate Kir2.1 localization. Specifically, we found that the Ldb19/Art1, Aly1/Art6, and Aly2/Art3 α-arrestin adaptor proteins promote Kir2.1 trafficking to the cell surface, increase Kir2.1 activity at the plasma membrane, and raise intracellular potassium levels. To better quantify the intracellular and cell-surface populations of Kir2.1, we created fluorogen-activating protein fusions and for the first time used this technique to measure the cell-surface residency of a plasma membrane protein in yeast. Our experiments revealed that two α-arrestin effectors also control Kir2.1 localization. In particular, both the Rsp5 ubiquitin ligase and the protein phosphatase calcineurin facilitated the α-arrestin-mediated trafficking of Kir2.1. Together, our findings implicate α-arrestins in regulating an additional class of plasma membrane proteins and establish a new tool for dissecting the trafficking itinerary of any membrane protein in yeast.


Assuntos
Arrestina/metabolismo , Membrana Celular/metabolismo , Endocitose/fisiologia , Modelos Biológicos , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Arrestina/genética , Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Transporte Proteico , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/genética
8.
J Vis Exp ; (123)2017 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-28570520

RESUMO

Filamentous actin (F-actin) organization within cells is regulated by a large number of actin-binding proteins that control actin nucleation, growth, cross-linking and/or disassembly. This protocol describes a technique - the actin co-sedimentation, or pelleting, assay - to determine whether a protein or protein domain binds F-actin and to measure the affinity of the interaction (i.e., the dissociation equilibrium constant). In this technique, a protein of interest is first incubated with F-actin in solution. Then, differential centrifugation is used to sediment the actin filaments, and the pelleted material is analyzed by SDS-PAGE. If the protein of interest binds F-actin, it will co-sediment with the actin filaments. The products of the binding reaction (i.e., F-actin and the protein of interest) can be quantified to determine the affinity of the interaction. The actin pelleting assay is a straightforward technique for determining if a protein of interest binds F-actin and for assessing how changes to that protein, such as ligand binding, affect its interaction with F-actin.


Assuntos
Citoesqueleto de Actina/química , Actinas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Ligação Proteica
9.
J Biol Chem ; 292(17): 7077-7086, 2017 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-28298447

RESUMO

Intercellular epithelial junctions formed by classical cadherins, ß-catenin, and the actin-binding protein α-catenin link the actin cytoskeletons of adjacent cells into a structural continuum. These assemblies transmit forces through the tissue and respond to intracellular and extracellular signals. However, the mechanisms of junctional assembly and regulation are poorly understood. Studies of cadherin-catenin assembly in a number of metazoans have revealed both similarities and unexpected differences in the biochemical properties of the cadherin·catenin complex that likely reflect the developmental and environmental requirements of different tissues and organisms. Here, we report the structural and biochemical characterization of HMP-1, the Caenorhabditis elegans α-catenin homolog, and compare it with mammalian α-catenin. HMP-1 shares overall similarity in structure and actin-binding properties, but displayed differences in conformational flexibility and allosteric regulation from mammalian α-catenin. HMP-1 bound filamentous actin with an affinity in the single micromolar range, even when complexed with the ß-catenin homolog HMP-2 or when present in a complex of HMP-2 and the cadherin homolog HMR-1, indicating that HMP-1 binding to F-actin is not allosterically regulated by the HMP-2·HMR-1 complex. The middle (i.e. M) domain of HMP-1 appeared to be less conformationally flexible than mammalian α-catenin, which may underlie the dampened effect of HMP-2 binding on HMP-1 actin-binding activity compared with that of the mammalian homolog. In conclusion, our data indicate that HMP-1 constitutively binds ß-catenin and F-actin, and although the overall structure and function of HMP-1 and related α-catenins are similar, the vertebrate proteins appear to be under more complex conformational regulation.


Assuntos
Actinas/química , Caderinas/química , Proteínas de Caenorhabditis elegans/química , Proteínas do Citoesqueleto/química , alfa Catenina/química , beta Catenina/química , Sítio Alostérico , Animais , Caenorhabditis elegans , Adesão Celular , Cristalografia por Raios X , Glutationa Transferase/metabolismo , Simulação de Dinâmica Molecular , Ligação Proteica , Domínios Proteicos , Coelhos , Relação Estrutura-Atividade , Vinculina/química
10.
J Biol Chem ; 291(30): 15687-99, 2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27231342

RESUMO

α-Catenin is the primary link between the cadherin·catenin complex and the actin cytoskeleton. Mammalian αE-catenin is allosterically regulated: the monomer binds the ß-catenin·cadherin complex, whereas the homodimer does not bind ß-catenin but interacts with F-actin. As part of the cadherin·catenin complex, αE-catenin requires force to bind F-actin strongly. It is not known whether these properties are conserved across the mammalian α-catenin family. Here we show that αT (testes)-catenin, a protein unique to amniotes that is expressed predominantly in the heart, is a constitutive actin-binding α-catenin. We demonstrate that αT-catenin is primarily a monomer in solution and that αT-catenin monomer binds F-actin in cosedimentation assays as strongly as αE-catenin homodimer. The ß-catenin·αT-catenin heterocomplex also binds F-actin with high affinity unlike the ß-catenin·αE-catenin complex, indicating that αT-catenin can directly link the cadherin·catenin complex to the actin cytoskeleton. Finally, we show that a mutation in αT-catenin linked to arrhythmogenic right ventricular cardiomyopathy, V94D, promotes homodimerization, blocks ß-catenin binding, and in cardiomyocytes disrupts localization at cell-cell contacts. Together, our data demonstrate that αT-catenin is a constitutively active actin-binding protein that can physically couple the cadherin·catenin complex to F-actin in the absence of tension. We speculate that these properties are optimized to meet the demands of cardiomyocyte adhesion.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Caderinas/metabolismo , Hipertrofia Ventricular Direita/metabolismo , Complexos Multiproteicos/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , alfa Catenina/metabolismo , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Actinas/química , Actinas/genética , Animais , Caderinas/química , Caderinas/genética , Hipertrofia Ventricular Direita/genética , Camundongos , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Miocárdio/patologia , Miócitos Cardíacos/patologia , Ligação Proteica , alfa Catenina/química , alfa Catenina/genética
11.
J Cell Biol ; 213(2): 261-74, 2016 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-27091449

RESUMO

Axons navigate long distances through complex 3D environments to interconnect the nervous system during development. Although the precise spatiotemporal effects of most axon guidance cues remain poorly characterized, a prevailing model posits that attractive guidance cues stimulate actin polymerization in neuronal growth cones whereas repulsive cues induce actin disassembly. Contrary to this model, we find that the repulsive guidance cue Slit stimulates the formation and elongation of actin-based filopodia from mouse dorsal root ganglion growth cones. Surprisingly, filopodia form and elongate toward sources of Slit, a response that we find is required for subsequent axonal repulsion away from Slit. Mechanistically, Slit evokes changes in filopodium dynamics by increasing direct binding of its receptor, Robo, to members of the actin-regulatory Ena/VASP family. Perturbing filopodium dynamics pharmacologically or genetically disrupts Slit-mediated repulsion and produces severe axon guidance defects in vivo. Thus, Slit locally stimulates directional filopodial extension, a process that is required for subsequent axonal repulsion downstream of the Robo receptor.


Assuntos
Axônios/metabolismo , Glicoproteínas/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Pseudópodes/fisiologia , Receptores Imunológicos/fisiologia , Animais , Axônios/ultraestrutura , Quimiotaxia , Desenvolvimento Embrionário , Glicoproteínas/metabolismo , Cones de Crescimento/metabolismo , Cones de Crescimento/ultraestrutura , Células HEK293 , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Pseudópodes/metabolismo , Pseudópodes/ultraestrutura , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Transdução de Sinais , Proteínas Roundabout
12.
PLoS Biol ; 11(11): e1001720, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24302887

RESUMO

Planar cell polarity (PCP) regulates cell alignment required for collective cell movement during embryonic development. This requires PCP/PCP effector proteins, some of which also play essential roles in ciliogenesis, highlighting the long-standing question of the role of the cilium in PCP. Wdpcp, a PCP effector, was recently shown to regulate both ciliogenesis and collective cell movement, but the underlying mechanism is unknown. Here we show Wdpcp can regulate PCP by direct modulation of the actin cytoskeleton. These studies were made possible by recovery of a Wdpcp mutant mouse model. Wdpcp-deficient mice exhibit phenotypes reminiscent of Bardet-Biedl/Meckel-Gruber ciliopathy syndromes, including cardiac outflow tract and cochlea defects associated with PCP perturbation. We observed Wdpcp is localized to the transition zone, and in Wdpcp-deficient cells, Sept2, Nphp1, and Mks1 were lost from the transition zone, indicating Wdpcp is required for recruitment of proteins essential for ciliogenesis. Wdpcp is also found in the cytoplasm, where it is localized in the actin cytoskeleton and in focal adhesions. Wdpcp interacts with Sept2 and is colocalized with Sept2 in actin filaments, but in Wdpcp-deficient cells, Sept2 was lost from the actin cytoskeleton, suggesting Wdpcp is required for Sept2 recruitment to actin filaments. Significantly, organization of the actin filaments and focal contacts were markedly changed in Wdpcp-deficient cells. This was associated with decreased membrane ruffling, failure to establish cell polarity, and loss of directional cell migration. These results suggest the PCP defects in Wdpcp mutants are not caused by loss of cilia, but by direct disruption of the actin cytoskeleton. Consistent with this, Wdpcp mutant cochlea has normal kinocilia and yet exhibits PCP defects. Together, these findings provide the first evidence, to our knowledge, that a PCP component required for ciliogenesis can directly modulate the actin cytoskeleton to regulate cell polarity and directional cell migration.


Assuntos
Citoesqueleto de Actina/metabolismo , Movimento Celular , Cílios/fisiologia , Proteínas do Citoesqueleto/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Polaridade Celular , Células Cultivadas , Análise Mutacional de DNA , Adesões Focais/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Transporte Proteico , Septinas/metabolismo , Imagem com Lapso de Tempo , Via de Sinalização Wnt , Peixe-Zebra
13.
Mol Biol Cell ; 24(23): 3710-20, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24068324

RESUMO

The actin-binding protein αE-catenin may contribute to transitions between cell migration and cell-cell adhesion that depend on remodeling the actin cytoskeleton, but the underlying mechanisms are unknown. We show that the αE-catenin actin-binding domain (ABD) binds cooperatively to individual actin filaments and that binding is accompanied by a conformational change in the actin protomer that affects filament structure. αE-catenin ABD binding limits barbed-end growth, especially in actin filament bundles. αE-catenin ABD inhibits actin filament branching by the Arp2/3 complex and severing by cofilin, both of which contact regions of the actin protomer that are structurally altered by αE-catenin ABD binding. In epithelial cells, there is little correlation between the distribution of αE-catenin and the Arp2/3 complex at developing cell-cell contacts. Our results indicate that αE-catenin binding to filamentous actin favors assembly of unbranched filament bundles that are protected from severing over more dynamic, branched filament arrays.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , alfa Catenina/química , alfa Catenina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Fatores de Despolimerização de Actina/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Animais , Microscopia Crioeletrônica , Cães , Processamento de Imagem Assistida por Computador , Células Madin Darby de Rim Canino , Camundongos , Modelos Moleculares , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
14.
J Biol Chem ; 288(31): 22324-32, 2013 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-23788645

RESUMO

It is unknown whether homologs of the cadherin·catenin complex have conserved structures and functions across the Metazoa. Mammalian αE-catenin is an allosterically regulated actin-binding protein that binds the cadherin·ß-catenin complex as a monomer and whose dimerization potentiates F-actin association. We tested whether these functional properties are conserved in another vertebrate, the zebrafish Danio rerio. Here we show, despite 90% sequence identity, that Danio rerio and Mus musculus αE-catenin have striking functional differences. We demonstrate that D. rerio αE-catenin is monomeric by size exclusion chromatography, native PAGE, and small angle x-ray scattering. D. rerio αE-catenin binds F-actin in cosedimentation assays as a monomer and as an α/ß-catenin heterodimer complex. D. rerio αE-catenin also bundles F-actin, as shown by negative stained transmission electron microscopy, and does not inhibit Arp2/3 complex-mediated actin nucleation in bulk polymerization assays. Thus, core properties of α-catenin function, F-actin and ß-catenin binding, are conserved between mouse and zebrafish. We speculate that unique regulatory properties have evolved to match specific developmental requirements.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas dos Microfilamentos/metabolismo , alfa Catenina/metabolismo , Animais , Cromatografia em Gel , Camundongos , Eletroforese em Gel de Poliacrilamida Nativa , Ligação Proteica , Espalhamento de Radiação , Peixe-Zebra
15.
Int Rev Cell Mol Biol ; 303: 1-25, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23445807

RESUMO

The regulated assembly and organization of actin filaments allows the cell to construct a large diversity of actin-based structures specifically suited to a range of cellular processes. A vast array of actin regulatory proteins must work in concert to form specific actin networks within cells, and spatial and temporal requirements for actin assembly necessitate rapid regulation of protein activity. This chapter explores a common mechanism of controlling the activity of actin binding proteins: allosteric autoinhibition by interdomain head-tail interactions. Intramolecular interactions maintain these proteins in a closed conformation that masks protein domains needed to regulate actin dynamics. Autoinhibition is typically relieved by two or more ligand binding and/or posttranslational modification events that expose key protein domains. Regulation through multiple inputs permits precise temporal and spatial control of protein activity to guide actin network formation.


Assuntos
Actinas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Regulação Alostérica , Animais , Humanos , Modelos Biológicos
16.
Proc Natl Acad Sci U S A ; 107(33): 14591-6, 2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20689042

RESUMO

The ternary complex of cadherin, beta-catenin, and alpha-catenin regulates actin-dependent cell-cell adhesion. alpha-Catenin can bind beta-catenin and F-actin, but in mammals alpha-catenin either binds beta-catenin as a monomer or F-actin as a homodimer. It is not known if this conformational regulation of alpha-catenin is evolutionarily conserved. The Caenorhabditis elegans alpha-catenin homolog HMP-1 is essential for actin-dependent epidermal enclosure and embryo elongation. Here we show that HMP-1 is a monomer with a functional C-terminal F-actin binding domain. However, neither full-length HMP-1 nor a ternary complex of HMP-1-HMP-2(beta-catenin)-HMR-1(cadherin) bind F-actin in vitro, suggesting that HMP-1 is auto-inhibited. Truncation of either the F-actin or HMP-2 binding domain of HMP-1 disrupts C. elegans development, indicating that HMP-1 must be able to bind F-actin and HMP-2 to function in vivo. Our study defines evolutionarily conserved properties of alpha-catenin and suggests that multiple mechanisms regulate alpha-catenin binding to F-actin.


Assuntos
Caderinas/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , alfa Catenina/metabolismo , Actinas/genética , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Caderinas/química , Caderinas/genética , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Eletroforese em Gel de Poliacrilamida , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Dados de Sequência Molecular , Mutação , Ligação Proteica , Multimerização Proteica , Espalhamento a Baixo Ângulo , Homologia de Sequência de Aminoácidos , Técnicas do Sistema de Duplo-Híbrido , Difração de Raios X , alfa Catenina/química , alfa Catenina/genética
17.
J Cell Biol ; 189(2): 339-52, 2010 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-20404114

RESUMO

alphaE-catenin binds the cell-cell adhesion complex of E-cadherin and beta-catenin (beta-cat) and regulates filamentous actin (F-actin) dynamics. In vitro, binding of alphaE-catenin to the E-cadherin-beta-cat complex lowers alphaE-catenin affinity for F-actin, and alphaE-catenin alone can bind F-actin and inhibit Arp2/3 complex-mediated actin polymerization. In cells, to test whether alphaE-catenin regulates actin dynamics independently of the cadherin complex, the cytosolic alphaE-catenin pool was sequestered to mitochondria without affecting overall levels of alphaE-catenin or the cadherin-catenin complex. Sequestering cytosolic alphaE-catenin to mitochondria alters lamellipodia architecture and increases membrane dynamics and cell migration without affecting cell-cell adhesion. In contrast, sequestration of cytosolic alphaE-catenin to the plasma membrane reduces membrane dynamics. These results demonstrate that the cytosolic pool of alphaE-catenin regulates actin dynamics independently of cell-cell adhesion.


Assuntos
Actinas/metabolismo , Caderinas/metabolismo , Adesão Celular/fisiologia , alfa Catenina/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Animais , Caderinas/genética , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Movimento Celular/fisiologia , Citoplasma/metabolismo , Cães , Humanos , Mitocôndrias/metabolismo , Multimerização Proteica , Pseudópodes/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , alfa Catenina/química , alfa Catenina/genética , Quinases da Família src/genética , Quinases da Família src/metabolismo
18.
J Neurosci ; 29(40): 12449-66, 2009 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-19812321

RESUMO

CNS synapse assembly typically follows after stable contacts between "appropriate" axonal and dendritic membranes are made. We show that presynaptic boutons selectively form de novo following neuronal fiber adhesion to beads coated with poly-d-lysine (PDL), an artificial cationic polypeptide. As demonstrated by atomic force and live confocal microscopy, functional presynaptic boutons self-assemble as rapidly as 1 h after bead contact, and are found to contain a variety of proteins characteristic of presynaptic endings. Interestingly, presynaptic compartment assembly does not depend on the presence of a biological postsynaptic membrane surface. Rather, heparan sulfate proteoglycans, including syndecan-2, as well as others possibly adsorbed onto the bead matrix or expressed on the axon surface, are required for assembly to proceed by a mechanism dependent on the dynamic reorganization of F-actin. Our results indicate that certain (but not all) nonspecific cationic molecules like PDL, with presumably electrostatically mediated adhesive properties, can effectively bypass cognate and natural postsynaptic ligands to trigger presynaptic assembly in the absence of specific target recognition. In contrast, we find that postsynaptic compartment assembly depends on the prior presence of a mature presynaptic ending.


Assuntos
Hipocampo/citologia , Hipocampo/metabolismo , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Animais , Axônios , Adesão Celular , Células Cultivadas , Hipocampo/embriologia , Proteínas de Membrana/metabolismo , Microscopia de Força Atômica , Microscopia Confocal , Ratos , Ratos Sprague-Dawley , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestrutura
19.
Mol Cell Neurosci ; 41(1): 44-50, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19386231

RESUMO

A complex program of cell intrinsic and extrinsic signals guide cortical development. Although genetic studies in mice have uncovered roles for numerous genes and gene families in multiple aspects of corticogenesis, determining their cell autonomous functions is often complicated by pleiotropic defects. Here we describe a novel lentiviral-based method to analyze cell autonomy by generating two-color chimeric mouse embryos. Ena/VASP-deficient mutant and control embryonic stem (ES) cells were labeled with different fluorescent chimeric proteins (EGFP and mCherry) that were modified to bind to the plasma membrane. These labeled ES cells were used to generate two-color chimeric embryos possessing two genetically distinct populations of cortical cells, permitting multiple aspects of neuronal morphogenesis to be analyzed and compared between the two cell populations. We observed little difference between the ability of control and Ena/VASP-deficient cells to contribute to cortical organization during development. In contrast, we observed axon fiber tracts originating from control neurons but not Ena/VASP-deficient neurons, indicating that loss of Ena/VASP causes a cell autonomous defect in cortical axon formation. This technique could be applied to determine other cell autonomous functions in different stages of cortical development.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Quimera , Neurônios , Sequência de Aminoácidos , Animais , Sequência de Bases , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Córtex Cerebral/metabolismo , Quimera/anatomia & histologia , Quimera/fisiologia , Corantes Fluorescentes/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Dados de Sequência Molecular , Morfogênese , Neurônios/citologia , Neurônios/fisiologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
20.
Nat Cell Biol ; 9(12): 1347-59, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18026093

RESUMO

Extension of neurites from a cell body is essential to form a functional nervous system; however, the mechanisms underlying neuritogenesis are poorly understood. Ena/VASP proteins regulate actin dynamics and modulate elaboration of cellular protrusions. We recently reported that cortical axon-tract formation is lost in Ena/VASP-null mice and Ena/VASP-null cortical neurons lack filopodia and fail to elaborate neurites. Here, we report that neuritogenesis in Ena/VASP-null neurons can be rescued by restoring filopodia formation through ectopic expression of the actin nucleating protein mDia2. Conversely, wild-type neurons in which filopodia formation is blocked fail to elaborate neurites. We also report that laminin, which promotes the formation of filopodia-like actin-rich protrusions, rescues neuritogenesis in Ena/VASP-deficient neurons. Therefore, filopodia formation is a key prerequisite for neuritogenesis in cortical neurons. Neurite initiation also requires microtubule extension into filopodia, suggesting that interactions between actin-filament bundles and dynamic microtubules within filopodia are crucial for neuritogenesis.


Assuntos
Córtex Cerebral/citologia , Neuritos/fisiologia , Neurônios/fisiologia , Pseudópodes/fisiologia , Actinas/metabolismo , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/fisiologia , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Laminina/fisiologia , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/fisiologia , Proteínas Associadas aos Microtúbulos , Microtúbulos/fisiologia , Mutação , Miosina Tipo II/antagonistas & inibidores , Miosinas/biossíntese , NADPH Desidrogenase/biossíntese , Neurônios/ultraestrutura , Fosfoproteínas/genética , Fosfoproteínas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...