Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Biol ; 216(7): 1925-1936, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28600434

RESUMO

Membrane remodeling plays a fundamental role during a variety of biological events. However, the dynamics and the molecular mechanisms regulating this process within cells in mammalian tissues in situ remain largely unknown. In this study, we use intravital subcellular microscopy in live mice to study the role of the actomyosin cytoskeleton in driving the remodeling of membranes of large secretory granules, which are integrated into the plasma membrane during regulated exocytosis. We show that two isoforms of nonmuscle myosin II, NMIIA and NMIIB, control distinct steps of the integration process. Furthermore, we find that F-actin is not essential for the recruitment of NMII to the secretory granules but plays a key role in the assembly and activation of NMII into contractile filaments. Our data support a dual role for the actomyosin cytoskeleton in providing the mechanical forces required to remodel the lipid bilayer and serving as a scaffold to recruit key regulatory molecules.


Assuntos
Células Acinares/metabolismo , Membrana Celular/metabolismo , Exocitose , Membranas Intracelulares/metabolismo , Fusão de Membrana , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Glândulas Salivares/metabolismo , Vesículas Secretórias/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Genótipo , Microscopia Intravital , Cinética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Microscopia de Vídeo , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIB/genética , Fenótipo , Glândulas Salivares/citologia , Transdução de Sinais
2.
Sci Rep ; 6: 24161, 2016 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-27063635

RESUMO

The actin cytoskeleton is a critical regulator of intestinal mucosal barrier permeability, and the integrity of epithelial adherens junctions (AJ) and tight junctions (TJ). Non muscle myosin II (NM II) is a key cytoskeletal motor that controls actin filament architecture and dynamics. While NM II has been implicated in the regulation of epithelial junctions in vitro, little is known about its roles in the intestinal mucosa in vivo. In this study, we generated a mouse model with an intestinal epithelial-specific knockout of NM IIA heavy chain (NM IIA cKO) and examined the structure and function of normal gut barrier, and the development of experimental colitis in these animals. Unchallenged NM IIA cKO mice showed increased intestinal permeability and altered expression/localization of several AJ/TJ proteins. They did not develop spontaneous colitis, but demonstrated signs of a low-scale mucosal inflammation manifested by prolapses, lymphoid aggregates, increased cytokine expression, and neutrophil infiltration in the gut. NM IIA cKO animals were characterized by a more severe disruption of the gut barrier and exaggerated mucosal injury during experimentally-induced colitis. Our study provides the first evidence that NM IIA plays important roles in establishing normal intestinal barrier, and protection from mucosal inflammation in vivo.


Assuntos
Mucosa Intestinal/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Claudinas/metabolismo , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Citocinas/metabolismo , Sulfato de Dextrana/toxicidade , Imunoglobulina A/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Neutrófilos/citologia , Neutrófilos/imunologia , Miosina não Muscular Tipo IIA/antagonistas & inibidores , Miosina não Muscular Tipo IIA/genética , Permeabilidade , Prolapso , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Junções Íntimas/metabolismo
3.
Sci Rep ; 5: 14068, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26369831

RESUMO

To investigate the contribution of nonmuscle myosin II-A (NM II-A) to early cardiac development we crossed Myh9 floxed mice and Nkx2.5 cre-recombinase mice. Nkx2.5 is expressed in the early heart (E7.5) and later in the tongue epithelium. Mice homozygous for deletion of NM II-A (A(Nkx)/A(Nkx)) are born at the expected ratio with normal hearts, but consistently develop an invasive squamous cell carcinoma (SCC) of the tongue (32/32 A(Nkx)/A(Nkx)) as early as E17.5. To assess reproducibility a second, independent line of Myh9 floxed mice derived from a different embryonic stem cell clone was tested. This second line also develops SCC indistinguishable from the first (15/15). In A(Nkx)/A(Nkx) mouse tongue epithelium, genetic deletion of NM II-A does not affect stabilization of TP53, unlike a previous report for SCC. We attribute the consistent, early formation of SCC with high penetrance to the role of NM II in maintaining mitotic stability during karyokinesis.


Assuntos
Carcinoma de Células Escamosas/genética , Transformação Celular Neoplásica/genética , Deleção de Genes , Miosina não Muscular Tipo IIA/genética , Neoplasias da Língua/genética , Animais , Carcinoma de Células Escamosas/patologia , Movimento Celular/genética , Modelos Animais de Doenças , Progressão da Doença , Expressão Gênica , Variação Genética , Genótipo , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Camundongos , Camundongos Knockout , Mucosa/metabolismo , Mucosa/patologia , Gradação de Tumores , Invasividade Neoplásica , Proteína Oncogênica p21(ras)/genética , Proteína Oncogênica p21(ras)/metabolismo , Fenótipo , Reprodutibilidade dos Testes , Neoplasias da Língua/patologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
4.
Hepatology ; 62(6): 1858-69, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26126491

RESUMO

UNLABELLED: Keratins, among other cytoskeletal intermediate filament proteins, are mutated at a highly conserved arginine with consequent severe disease phenotypes due to disruption of keratin filament organization. We screened a kinase inhibitor library, using A549 cells that are transduced with a lentivirus keratin 18 (K18) construct, to identify compounds that normalize filament disruption due to K18 Arg90Cys mutation at the conserved arginine. High-throughput screening showed that PKC412, a multikinase inhibitor, ameliorated K18 Arg90Cys-mediated keratin filament disruption in cells and in the livers of previously described transgenic mice that overexpress K18 Arg90Cys. Furthermore, PKC412 protected cultured A549 cells that express mutant or wild-type K18 and mouse livers of the K18 Arg90Cys-overexpressing transgenic mice from Fas-induced apoptosis. Proteomic analysis of proteins that associated with keratins after exposure of K18-expressing A549 cells to PKC412 showed that nonmuscle myosin heavy chain-IIA (NMHC-IIA) partitions with the keratin fraction. The nonmuscle myosin-IIA (NM-IIA) association with keratins was confirmed by immune staining and by coimmunoprecipitation. The keratin-myosin association is myosin dephosphorylation-dependent; occurs with K8, the obligate K18 partner; is enhanced by PKC412 in cells and mouse liver; and is blocked by hyperphosphorylation conditions in cultured cells and mouse liver. Furthermore, NMHC-IIA knockdown inhibits PKC412-mediated normalization of K18 R90C filaments. CONCLUSION: The inhibitor PKC412 normalizes K18 Arg90Cys mutation-induced filament disruption and disorganization by enhancing keratin association with NM-IIA in a myosin dephosphorylation-regulated manner. Targeting of intermediate filament disorganization by compounds that alter keratin interaction with their associated proteins offers a potential novel therapeutic approach for keratin and possibly other intermediate filament protein-associated diseases.


Assuntos
Filamentos Intermediários/genética , Queratinas/metabolismo , Hepatopatias/genética , Mutação , Miosinas/metabolismo , Estaurosporina/análogos & derivados , Animais , Camundongos , Camundongos Transgênicos , Ligação Proteica , Estaurosporina/fisiologia
5.
J Am Soc Nephrol ; 26(5): 1081-91, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25168025

RESUMO

The kidney develops from reciprocal interactions between the metanephric mesenchyme and ureteric bud. The mesenchyme transforms into epithelia and forms complicated nephron structures, whereas the ureteric bud extends its pre-existing epithelial ducts. Although the roles are well established for extracellular stimuli, such as Wnt and Notch, it is unclear how the intracellular cytoskeleton regulates these morphogenetic processes. Myh9 and Myh10 encode nonmuscle myosin II heavy chains, and Myh9 mutations in humans are implicated in congenital kidney diseases and focal segmental glomerulosclerosis in adults. Here, we analyzed the roles of Myh9 and Myh10 in the developing kidney. Ureteric bud-specific depletion of Myh9 resulted in no apparent phenotypes, whereas mesenchyme-specific Myh9 deletion caused proximal tubule dilations and renal failure. Mesenchyme-specific Myh9/Myh10 mutant mice died shortly after birth and showed a severe defect in nephron formation. The nascent mutant nephrons failed to form a continuous lumen, which likely resulted from impaired apical constriction of the elongating tubules. In addition, nephron progenitors lacking Myh9/Myh10 or the possible interactor Kif26b were less condensed at midgestation and reduced at birth. Taken together, nonmuscle myosin II regulates the morphogenesis of immature nephrons derived from the metanephric mesenchyme and the maintenance of nephron progenitors. Our data also suggest that Myh9 deletion in mice results in failure to maintain renal tubules but not in glomerulosclerosis.


Assuntos
Morfogênese , Cadeias Pesadas de Miosina/fisiologia , Néfrons/embriologia , Miosina não Muscular Tipo IIA/fisiologia , Miosina não Muscular Tipo IIB/fisiologia , Animais , Animais Recém-Nascidos , Mesoderma/fisiologia , Camundongos Endogâmicos C57BL , Néfrons/metabolismo , Isoformas de Proteínas/metabolismo
6.
Dev Biol ; 382(1): 136-48, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23911870

RESUMO

In studies initially focused on roles of nonmuscle myosin IIA (NMIIA) in the developing mouse epidermis, we have discovered that a previously described cytokeratin 5 (K5)-Cre gene construct is expressed in early embryo development. Mice carrying floxed alleles of the nonmuscle myosin II heavy chain gene (NMHC IIA(flox/flox)) were crossed with the K5-Cre line. The progeny of newborn pups did not show a Mendelian genotype distribution, suggesting embryonic lethality. Analysis of post-implantation conceptuses from embryonic day (E)9.5 to E13.5 revealed poorly developed embryos and defective placentas, with significantly reduced labyrinth surface area and blood vessel vascularization. These results suggested the novel possibility that the bovine K5 promoter-driven Cre-recombinase was active early in trophoblast-lineage cells that give rise to the placenta. To test this possibility, K5-Cre transgenic mice were crossed with the mT/mG reporter mouse in which activation of GFP expression indicates Cre transgene expression. We observed activation of K5-Cre-driven GFP expression in the ectoplacental cone, in the extraembryonic ectoderm, and in trophoblast giant cells in the E6.5 embryo. In addition, we observed GFP expression at E11.5 to E13.5 in both the labyrinth of the placenta and the yolk sac. NMIIA expression was detected in these same cell types in normal embryos, as well as in E13.5 yolk sac and labyrinth. These findings taken together suggest that NMHC IIA may play critical roles in the early trophoblast-derived ectoplacental cone and extraembryonic ectoderm, as well as in the yolk sac and labyrinth tissues that form later. Our findings are consistent with phenotypes of constitutive NMIIA knockout mice made earlier, that displayed labyrinth and yolk sac-specific defects, but our findings extend those observations by suggesting possible NMIIA roles in trophoblast lineages as well. These results furthermore demonstrate that K5-Cre gene constructs, previously reported to be activated starting at approximately E12.5 in the forming epidermis, may be widely useful as drivers for activation of cre/lox based gene excision in early embryo extraembronic trophoblast tissues as well.


Assuntos
Ectoderma/embriologia , Perda do Embrião/patologia , Integrases/metabolismo , Queratina-5/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Trofoblastos/metabolismo , Trofoblastos/patologia , Alelos , Animais , Animais Recém-Nascidos , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Bovinos , Linhagem da Célula , Proliferação de Células , Cruzamentos Genéticos , Ectoderma/metabolismo , Ectoderma/patologia , Perda do Embrião/metabolismo , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Desenvolvimento Embrionário , Epiderme/embriologia , Epiderme/metabolismo , Epiderme/patologia , Feminino , Deleção de Genes , Genótipo , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Knockout , Gravidez , Saco Vitelino/metabolismo
7.
Curr Biol ; 23(8): 731-6, 2013 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-23562268

RESUMO

Nonmuscle myosin II (NMII) is thought to be the master integrator of force within epithelial apical junctions, mediating epithelial tissue morphogenesis and tensional homeostasis. Mutations in NMII are associated with a number of diseases due to failures in cell-cell adhesion. However, the organization and the precise mechanism by which NMII generates and responds to tension along the intercellular junctional line are still not known. We discovered that periodic assemblies of bipolar NMII filaments interlace with perijunctional actin and α-actinin to form a continuous belt of muscle-like sarcomeric units (∼400-600 nm) around each epithelial cell. Remarkably, the sarcomeres of adjacent cells are precisely paired across the junctional line, forming an integrated, transcellular contractile network. The contraction/relaxation of paired sarcomeres concomitantly impacts changes in apical cell shape and tissue geometry. We show differential distribution of NMII isoforms across heterotypic junctions and evidence for compensation between isoforms. Our results provide a model for how NMII force generation is effected along the junctional perimeter of each cell and communicated across neighboring cells in the epithelial organization. The sarcomeric network also provides a well-defined target to investigate the multiple roles of NMII in junctional homeostasis as well as in development and disease.


Assuntos
Junções Intercelulares/metabolismo , Miosina Tipo II/metabolismo , Órgão Espiral/metabolismo , Citoesqueleto de Actina/metabolismo , Actinina/metabolismo , Actinas/metabolismo , Animais , Compostos Heterocíclicos de 4 ou mais Anéis/metabolismo , Camundongos , Microscopia de Fluorescência , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo II/genética , Ratos
8.
J Biol Chem ; 287(33): 27345-58, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22718763

RESUMO

Ligand-induced internalization of the epidermal growth factor receptor (EGFR) is an important process for regulating signal transduction, cellular dynamics, and cell-cell communication. Here, we demonstrate that nonmuscle myosin II (NM II) is required for the internalization of the EGFR and to trigger the EGFR-dependent activation of ERK and AKT. The EGFR was identified as a protein that interacts with NM II by co-immunoprecipitation and mass spectrometry analysis. This interaction requires both the regulatory light chain 20 (RLC20) of NM II and the kinase domain of the EGFR. Two paralogs of NM II, NM II-A, and NM II-B can act to internalize the EGFR, depending on the cell type and paralog content of the cell line. Loss (siRNA) or inhibition (25 µm blebbistatin) of NM II attenuates the internalization of the EGFR and impairs EGFR-dependent activation of ERK and AKT. Both internalization of the EGFR and downstream signaling to ERK and AKT can be partially restored in siRNA-treated cells by introduction of wild type (WT) GFP-NM II, but cannot be restored by motor mutant NM II. Taken together, these results suggest that NM II plays a role in the internalization of the EGFR and EGFR-mediated signaling pathways.


Assuntos
Receptores ErbB/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Transdução de Sinais/fisiologia , Animais , Células COS , Chlorocebus aethiops , Ativação Enzimática/fisiologia , Receptores ErbB/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Humanos , Espectrometria de Massas , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIB/genética , Transporte Proteico/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
9.
Proc Natl Acad Sci U S A ; 109(12): 4509-14, 2012 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-22393000

RESUMO

During vertebrate cytokinesis it is thought that contractile ring constriction is driven by nonmuscle myosin II (NM II) translocation of antiparallel actin filaments. Here we report in situ, in vitro, and in vivo observations that challenge this hypothesis. Graded knockdown of NM II in cultured COS-7 cells reveals that the amount of NM II limits ring constriction. Restoration of the constriction rate with motor-impaired NM II mutants shows that the ability of NM II to translocate actin is not required for cytokinesis. Blebbistatin inhibition of cytokinesis indicates the importance of myosin strongly binding to actin and exerting tension during cytokinesis. This role is substantiated by transient kinetic experiments showing that the load-dependent mechanochemical properties of mutant NM II support efficient tension maintenance despite the inability to translocate actin. Under loaded conditions, mutant NM II exhibits a prolonged actin attachment in which a single mechanoenzymatic cycle spans most of the time of cytokinesis. This prolonged attachment promotes simultaneous binding of NM II heads to actin, thereby increasing tension and resisting expansion of the ring. The detachment of mutant NM II heads from actin is enhanced by assisting loads, which prevent mutant NM II from hampering furrow ingression during cytokinesis. In the 3D context of mouse hearts, mutant NM II-B R709C that cannot translocate actin filaments can rescue multinucleation in NM II-B ablated cardiomyocytes. We propose that the major roles of NM II in vertebrate cell cytokinesis are to bind and cross-link actin filaments and to exert tension on actin during contractile ring constriction.


Assuntos
Actinas/metabolismo , Miosina Tipo II/metabolismo , Actinas/química , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Reagentes de Ligações Cruzadas/química , Citocinese , Cinética , Camundongos , Microscopia de Fluorescência/métodos , Contração Muscular , Mutação , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Transporte Proteico , Fatores de Tempo
10.
J Cell Sci ; 125(Pt 9): 2244-56, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22328520

RESUMO

Recent evidence suggests that organization of the extracellular matrix (ECM) into aligned fibrils or fibril-like ECM topographies promotes rapid migration in fibroblasts. However, the mechanisms of cell migration that are altered by these changes in micro-environmental topography remain unknown. Here, using 1D fibrillar migration as a model system for oriented fibrillar 3D matrices, we find that fibroblast leading-edge dynamics are enhanced by 1D fibrillar micropatterns and demonstrate a dependence on the spatial positioning of cell adhesions. Although 1D, 2D and 3D matrix adhesions have similar assembly kinetics, both 1D and 3D adhesions are stabilized for prolonged periods, whereas both paxillin and vinculin show slower turnover rates in 1D adhesions. Moreover, actin in 1D adhesions undergoes slower retrograde flow than the actin that is present in 2D lamellipodia. These data suggest an increase in mechanical coupling between adhesions and protrusive machinery. Experimental reduction of contractility resulted in the loss of 1D adhesion structure and stability, with scattered small and unstable adhesions, and an uncoupling of adhesion protein-integrin stability. Genetic ablation of myosin IIA (MIIA) or myosin IIB (MIIB) isoforms revealed that MIIA is required for efficient migration in restricted environments as well as adhesion maturation, whereas MIIB helps to stabilize adhesions beneath the cell body. These data suggest that restricted cell environments, such as 1D patterns, require cellular contraction through MIIA to enhance adhesion stability and coupling to integrins behind the leading edge. This increase in mechanical coupling allows for greater leading-edge protrusion and rapid cell migration.


Assuntos
Microambiente Celular/fisiologia , Fibroblastos/fisiologia , Miosina não Muscular Tipo IIA/antagonistas & inibidores , Miosina não Muscular Tipo IIB/antagonistas & inibidores , Pseudópodes/fisiologia , Actinas/metabolismo , Animais , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Matriz Extracelular/metabolismo , Técnicas de Inativação de Genes , Integrinas/metabolismo , Camundongos , Células NIH 3T3 , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIB/genética , Paxilina/metabolismo , Plasmídeos , Transfecção , Vinculina/metabolismo
11.
Blood ; 119(1): 238-50, 2012 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-21908426

RESUMO

We have generated 3 mouse lines, each with a different mutation in the nonmuscle myosin II-A gene, Myh9 (R702C, D1424N, and E1841K). Each line develops MYH9-related disease similar to that found in human patients. R702C mutant human cDNA fused with green fluorescent protein was introduced into the first coding exon of Myh9, and D1424N and E1841K mutations were introduced directly into the corresponding exons. Homozygous R702C mice die at embryonic day 10.5-11.5, whereas homozygous D1424N and E1841K mice are viable. All heterozygous and homozygous mutant mice show macrothrombocytopenia with prolonged bleeding times, a defect in clot retraction, and increased extramedullary megakaryocytes. Studies of cultured megakaryocytes and live-cell imaging of megakaryocytes in the BM show that heterozygous R702C megakaryocytes form fewer and shorter proplatelets with less branching and larger buds. The results indicate that disrupted proplatelet formation contributes to the macrothrombocytopenia in mice and most probably in humans. We also observed premature cataract formation, kidney abnormalities, including albuminuria, focal segmental glomerulosclerosis and progressive kidney disease, and mild hearing loss. Our results show that heterozygous mice with mutations in the myosin motor or filament-forming domain manifest similar hematologic, eye, and kidney phenotypes to humans with MYH9-related disease.


Assuntos
Catarata/etiologia , Modelos Animais de Doenças , Perda Auditiva/etiologia , Nefropatias/etiologia , Megacariócitos/patologia , Mutação/genética , Miosina não Muscular Tipo IIA/fisiologia , Trombocitopenia/etiologia , Animais , Catarata/metabolismo , Catarata/patologia , Feminino , Imunofluorescência , Genes Letais , Perda Auditiva/metabolismo , Perda Auditiva/patologia , Heterozigoto , Homozigoto , Humanos , Immunoblotting , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Megacariócitos/metabolismo , Camundongos , Camundongos Transgênicos , Cadeias Pesadas de Miosina , Contagem de Plaquetas , Trombocitopenia/metabolismo , Trombocitopenia/patologia
12.
Biochem Soc Trans ; 39(5): 1131-5, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21936777

RESUMO

We propose that the in vivo functions of NM II (non-muscle myosin II) can be divided between those that depend on the N-terminal globular motor domain and those less dependent on motor activity but more dependent on the C-terminal domain. The former, being more dependent on the kinetic properties of NM II to translocate actin filaments, are less amenable to substitution by different NM II isoforms, whereas the in vivo functions of the latter, which involve the structural properties of NM II to cross-link actin filaments, are more amenable to substitution. In light of this hypothesis, we examine the ability of NM II-A, as well as a motor-compromised form of NM II-B, to replace NM II-B and rescue neuroepithelial cell-cell adhesion defects and hydrocephalus in the brain of NM II-B-depleted mice. We also examine the ability of NM II-B as well as chimaeric forms of NM II (II-A head and II-B tail and vice versa) to substitute for NM II-A in cell-cell adhesions in II-A-ablated mice. However, we also show that certain functions, such as neuronal cell migration in the developing brain and vascularization of the mouse embryo and placenta, specifically require NM II-B and II-A respectively.


Assuntos
Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Isoformas de Proteínas/metabolismo , Actinas/metabolismo , Animais , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Feminino , Camundongos , Neurônios/citologia , Neurônios/fisiologia , Miosina não Muscular Tipo IIA/química , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIB/química , Miosina não Muscular Tipo IIB/genética , Placenta/citologia , Placenta/patologia , Placenta/fisiologia , Gravidez , Isoformas de Proteínas/química , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
13.
Nat Immunol ; 11(10): 953-61, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20835229

RESUMO

During trafficking through tissues, T cells fine-tune their motility to balance the extent and duration of cell-surface contacts versus the need to traverse an entire organ. Here we show that in vivo, myosin IIA-deficient T cells had a triad of defects, including overadherence to high-endothelial venules, less interstitial migration and inefficient completion of recirculation through lymph nodes. Spatiotemporal analysis of three-dimensional motility in microchannels showed that the degree of confinement and myosin IIA function, rather than integrin adhesion (as proposed by the haptokinetic model), optimized motility rate. This motility occurred via a myosin IIA-dependent rapid 'walking' mode with multiple small and simultaneous adhesions to the substrate, which prevented spurious and prolonged adhesions. Adhesion discrimination provided by myosin IIA is thus necessary for the optimization of motility through complex tissues.


Assuntos
Adesão Celular/fisiologia , Movimento Celular , Linfonodos/imunologia , Miosina não Muscular Tipo IIA/fisiologia , Linfócitos T/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
Nat Commun ; 1: 71, 2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20842192

RESUMO

Human pluripotent stem (hPS) cells such as human embryonic stem (hES) and induced pluripotent stem (hiPS) cells are vulnerable under single cell conditions, which hampers practical applications; yet, the mechanisms underlying this cell death remain elusive. In this paper, we demonstrate that treatment with a specific inhibitor of non-muscle myosin II (NMII), blebbistatin, enhances the survival of hPS cells under clonal density and suspension conditions, and, in combination with a synthetic matrix, supports a fully defined environment for self-renewal. Consistent with this, genetically engineered mouse embryonic stem cells lacking an isoform of NMII heavy chain (NMHCII), or hES cells expressing a short hairpin RNA to knock down NMHCII, show greater viability than controls. Moreover, NMII inhibition increases the expression of self-renewal regulators Oct3/4 and Nanog, suggesting a mechanistic connection between NMII and self-renewal. These results underscore the importance of the molecular motor, NMII, as a novel target for chemically engineering the survival and self-renewal of hPS cells.


Assuntos
Miosina Tipo II/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Vetores Genéticos/genética , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Marcação In Situ das Extremidades Cortadas , Cariotipagem , Lentivirus/genética , Camundongos , Miosina Tipo II/genética , Células-Tronco Pluripotentes/efeitos dos fármacos , RNA Interferente Pequeno/genética
15.
Mol Biol Cell ; 21(22): 3952-62, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20861308

RESUMO

Ablation of nonmuscle myosin (NM) II-A or NM II-B results in mouse embryonic lethality. Here, we report the results of ablating NM II-C as well as NM II-C/II-B together in mice. NM II-C ablated mice survive to adulthood and show no obvious defects compared with wild-type littermates. However, ablation of NM II-C in mice expressing only 12% of wild-type amounts of NM II-B results in a marked increase in cardiac myocyte hypertrophy compared with the NM II-B hypomorphic mice alone. In addition, these hearts develop interstitial fibrosis associated with diffuse N-cadherin and ß-catenin localization at the intercalated discs, where both NM II-B and II-C are normally concentrated. When both NM II-C and II-B are ablated the B-C-/B-C- cardiac myocytes show major defects in karyokinesis. More than 90% of B-C-/B-C- myocytes demonstrate defects in chromatid segregation and mitotic spindle formation accompanied by increased stability of microtubules and abnormal formation of multiple centrosomes. This requirement for NM II in karyokinesis is further demonstrated in the HL-1 cell line derived from mouse atrial myocytes, by using small interfering RNA knockdown of NM II or treatment with the myosin inhibitor blebbistatin. Our study shows that NM II is involved in regulating cardiac myocyte karyokinesis by affecting microtubule dynamics.


Assuntos
Divisão do Núcleo Celular/fisiologia , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo II/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Animais , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Divisão do Núcleo Celular/genética , Chlorocebus aethiops , Feminino , Células HT29 , Células HeLa , Coração/embriologia , Coração/crescimento & desenvolvimento , Humanos , Pulmão/embriologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microtúbulos/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Cadeias Pesadas de Miosina/genética , Miosina Tipo II/genética , Miosina não Muscular Tipo IIB/genética , Interferência de RNA
16.
Proc Natl Acad Sci U S A ; 107(33): 14645-50, 2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20679233

RESUMO

Nonmuscle myosins (NMs) II-A and II-B are essential for embryonic mouse development, but their specific roles are not completely defined. Here we examine the isoforms and their domain specifically in vivo and in vitro by studying mice and cells in which nonmuscle myosin heavy chain (NMHC) II-A is genetically replaced by NMHC II-B or chimeric NMHC IIs that exchange the rod and head domains of NM II-A and II-B. In contrast with the failure of visceral endoderm formation resulting in embryonic day (E)6.5 lethality of A(-)/A(-) mice, replacement with NM II-B or chimeric NM IIs restores a normal visceral endoderm. This finding is consistent with NM II's role in cell adhesion and also confirms an essential, isoform-independent requirement for NM II in visceral endoderm function. The knock-in mice die between E9.5 and 12.5 because of defects in placenta formation associated with abnormal angiogenesis and cell migration, revealing a unique function for NM II-A in placenta development. In vitro results further support a requirement for NM II-A in directed cell migration and focal adhesion formation. These findings demonstrate an isoform-specific role for NM II-A during these processes, making replacement by another isoform, or chimeric NM II isoforms, less successful. The failure of these substitutions is not only related to the different kinetic properties of NM II-A and II-B, but also to their subcellular localization determined by the C-terminal domain. These results highlight the functions of the N-terminal motor and C-terminal rod domains of NM II and their different roles in cell-cell and cell-matrix adhesion.


Assuntos
Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Animais , Sítios de Ligação , Adesão Celular , Comunicação Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Feminino , Fibroblastos/citologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Microscopia Confocal , Mutação , Cadeias Pesadas de Miosina/genética , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIB/genética , Placenta/embriologia , Placenta/metabolismo , Gravidez , Fatores de Tempo
17.
Nat Cell Biol ; 12(7): 696-702, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20543839

RESUMO

Classic cadherin receptors cooperate with regulators of the actin cytoskeleton to control tissue organization in health and disease. At the apical junctions of epithelial cells, the cadherin ring of the zonula adherens (ZA) couples with a contiguous ring of actin filaments to support morphogenetic processes such as tissue integration and cellular morphology. However, the molecular mechanisms that coordinate adhesion and cytoskeleton at these junctions are poorly understood. Previously we identified non-muscle myosin II as a target of Rho signalling that supports cadherin junctions in mammalian epithelial cells. Myosin II has various cellular functions, which are increasingly attributable to the specific biophysical properties and regulation of its different isoforms. Here we report that myosin II isoforms have distinct and necessary roles at cadherin junctions. Although two of the three mammalian myosin II isoforms are found at the ZA, their localization is regulated by different upstream signalling pathways. Junctional localization of myosin IIA required E-cadherin adhesion, Rho/ROCK and myosin light-chain kinase, whereas junctional myosin IIB depended on Rap1. Further, these myosin II isoforms support E-cadherin junction integrity by different mechanisms. Myosin IIA RNA-mediated interference (RNAi) selectively perturbed the accumulation of E-cadherin in the apical ZA, decreased cadherin homophilic adhesion and disrupted cadherin clustering. In contrast, myosin IIB RNAi decreased filament content, altered dynamics, and increased the lateral movement of the perijunctional actin ring. Myosin IIA and IIB therefore identify two distinct functional modules, with different upstream signals that control junctional localization, and distinct functional effects. We propose that these two isoform-based modules cooperate to coordinate adhesion receptor and F-actin organization to form apical cadherin junctions.


Assuntos
Junções Aderentes/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Humanos , Modelos Biológicos
18.
Am J Pathol ; 174(2): 436-48, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19147824

RESUMO

Epithelial cell motility is critical for self-rejuvenation of normal intestinal mucosa, wound repair, and cancer metastasis. This process is regulated by the reorganization of the F-actin cytoskeleton, which is driven by a myosin II motor. However, the role of myosin II in regulating epithelial cell migration remains poorly understood. This study addressed the role of non-muscle myosin (NM) IIA in two different modes of epithelial cell migration: two-dimensional (2-D) migration that occurs during wound closure and three-dimensional (3-D) migration through a Matrigel matrix that occurs during cancer metastasis. Pharmacological inhibition or siRNA-mediated knockdown of NM IIA in SK-CO15 human colonic epithelial cells resulted in decreased 2-D migration and increased 3-D invasion. The attenuated 2-D migration was associated with increased cell adhesiveness to collagen and laminin and enhanced expression of beta1-integrin and paxillin. On the 2-D surface, NM IIA-deficient SK-CO15 cells failed to assemble focal adhesions and F-actin stress fibers. In contrast, the enhanced invasion of NM IIA-depleted cells was dependent on Raf-ERK1/2 signaling pathway activation, enhanced calpain activity, and increased calpain-2 expression. Our findings suggest that NM IIA promotes 2-D epithelial cell migration but antagonizes 3-D invasion. These observations indicate multiple functions for NM IIA, which, along with the regulation of the F-actin cytoskeleton and cell-matrix adhesions, involve previously unrecognized control of intracellular signaling and protein expression.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Mucosa Intestinal/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Colágeno , Combinação de Medicamentos , Imunofluorescência , Humanos , Imuno-Histoquímica , Laminina , Microscopia Confocal , Proteoglicanas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cicatrização/fisiologia
19.
Mol Biol Cell ; 19(12): 5032-46, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18799623

RESUMO

Rap1 enhances integrin-mediated adhesion but the link between Rap1 activation and integrin function in collagen phagocytosis is not defined. Mass spectrometry of Rap1 immunoprecipitates showed that the association of Rap1 with nonmuscle myosin heavy-chain II-A (NMHC II-A) was enhanced by cell attachment to collagen beads. Rap1 colocalized with NM II-A at collagen bead-binding sites. There was a transient increase in myosin light-chain phosphorylation after collagen-bead binding that was dependent on myosin light-chain kinase but not Rho kinase. Inhibition of myosin light-chain phosphorylation, but not myosin II-A motor activity inhibited collagen-bead binding and Rap activation. In vitro binding assays demonstrated binding of Rap1A to filamentous myosin rods, and in situ staining of permeabilized cells showed that NM II-A filaments colocalized with F-actin at collagen bead sites. Knockdown of NM II-A did not affect talin, actin, or beta1-integrin targeting to collagen beads but targeting of Rap1 and vinculin to collagen was inhibited. Conversely, knockdown of Rap1 did not affect localization of NM II-A to beads. We conclude that MLC phosphorylation in response to initial collagen-bead binding promotes NM II-A filament assembly; binding of Rap1 to myosin filaments enables Rap1-dependent integrin activation and enhanced collagen phagocytosis.


Assuntos
Colágeno/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Fagocitose/fisiologia , Proteínas rap1 de Ligação ao GTP/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/fisiologia , Compostos Heterocíclicos de 4 ou mais Anéis/metabolismo , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Camundongos , Dados de Sequência Molecular , Quinase de Cadeia Leve de Miosina/genética , Quinase de Cadeia Leve de Miosina/metabolismo , Miosina não Muscular Tipo IIA/genética , Peptídeos/genética , Peptídeos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Talina/genética , Talina/metabolismo , Proteínas rap1 de Ligação ao GTP/genética
20.
J Cell Sci ; 121(Pt 1): 11-8, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18096687

RESUMO

For many years, analyses of the role of the actomyosin cytoskeleton in many basic cellular processes have centered on actin. Increasingly, however, a number of investigators are examining proteins that are proximal to actin; in particular, nonmuscle myosin II (NMII). Recent experiments have increased our understanding of the role of NMII in three related cellular activities: generation of cell polarity, cell migration and cell-cell adhesion. Progress has been particularly promising thanks to the use of new microscopic, genetic and biochemical techniques. In mammalian systems, generation of transgenic mice and the introduction of specific siRNAs have been useful in deciphering the role of the three different isoforms of NMII: NMIIA, NMIIB and NMIIC. Studies in Drosophila and Aplysia, which are informative model systems for investigating the function of NMII, have also shed light on NMII. Recent work examines the contractile and structural roles that NMII plays at cell-cell boundaries, and both its contractile and actin-crosslinking roles in cell migration. In addition, NMII might also function as a scaffold molecule, anchoring signaling molecules, such as kinases and Rho GTPase guanine nucleotide exchange factors.


Assuntos
Movimento Celular , Miosina Tipo II/fisiologia , Animais , Adesão Celular , Polaridade Celular , Gastrulação , Humanos , Miosina Tipo II/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...