Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Dev Biol ; 470: 49-61, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33188738

RESUMO

Mutations in non-muscle myosin 2A (NM2A) encompass a wide spectrum of anomalies collectively known as MYH9-Related Disease (MYH9-RD) in humans that can include macrothrombocytopenia, glomerulosclerosis, deafness, and cataracts. We previously created mouse models of the three mutations most frequently found in humans: R702C, D1424N, and E1841K. While homozygous R702C and D1424N mutations are embryonic lethal, we found homozygous mutant E1841K mice to be viable. However the homozygous male, but not female, mice were infertile. Here, we report that these mice have reduced testis size and defects in actin-associated junctions in Sertoli cells, resulting in inability to form the blood-testis barrier and premature germ cell loss. Moreover, compound double heterozygous (R702C/E1841K and D1424/E1841K) males show the same abnormalities in testes as E1841K homozygous males. Conditional ablation of either NM2A or NM2B alone in Sertoli cells has no effect on fertility and testis size, however deletion of both NM2A and NM2B in Sertoli cells results in infertility. Isolation of mutant E1841K Sertoli cells reveals decreased NM2A and F-actin colocalization and thicker NM2A filaments. Furthermore, AE1841K/AE1841K and double knockout Sertoli cells demonstrate microtubule disorganization and increased tubulin acetylation, suggesting defects in the microtubule cytoskeleton. Together, these results demonstrate that NM2A and 2B paralogs play redundant roles in Sertoli cells and are essential for testes development and normal fertility.


Assuntos
Actomiosina/metabolismo , Citoesqueleto/ultraestrutura , Infertilidade Masculina/genética , Cadeias Pesadas de Miosina/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Células de Sertoli/fisiologia , Actinas/metabolismo , Actomiosina/química , Animais , Barreira Hematotesticular/metabolismo , Forma Celular , Citoesqueleto/metabolismo , Infertilidade Masculina/patologia , Infertilidade Masculina/fisiopatologia , Masculino , Camundongos , Microtúbulos/química , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Cadeias Pesadas de Miosina/genética , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIB/genética , Miosina não Muscular Tipo IIB/metabolismo , Tamanho do Órgão , Permeabilidade , Mutação Puntual , Células de Sertoli/citologia , Células de Sertoli/ultraestrutura , Testículo/patologia , Tubulina (Proteína)/metabolismo
2.
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
3.
Nat Rev Mol Cell Biol ; 10(11): 778-90, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19851336

RESUMO

Non-muscle myosin II (NM II) is an actin-binding protein that has actin cross-linking and contractile properties and is regulated by the phosphorylation of its light and heavy chains. The three mammalian NM II isoforms have both overlapping and unique properties. Owing to its position downstream of convergent signalling pathways, NM II is central in the control of cell adhesion, cell migration and tissue architecture. Recent insight into the role of NM II in these processes has been gained from loss-of-function and mutant approaches, methods that quantitatively measure actin and adhesion dynamics and the discovery of NM II mutations that cause monogenic diseases.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Miosina Tipo II/metabolismo , Animais , Humanos
4.
J Cell Sci ; 131(6)2018 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-29487177

RESUMO

Many actin filaments in animal cells are co-polymers of actin and tropomyosin. In many cases, non-muscle myosin II associates with these co-polymers to establish a contractile network. However, the temporal relationship of these three proteins in the de novo assembly of actin filaments is not known. Intravital subcellular microscopy of secretory granule exocytosis allows the visualisation and quantification of the formation of an actin scaffold in real time, with the added advantage that it occurs in a living mammal under physiological conditions. We used this model system to investigate the de novo assembly of actin, tropomyosin Tpm3.1 (a short isoform of TPM3) and myosin IIA (the form of non-muscle myosin II with its heavy chain encoded by Myh9) on secretory granules in mouse salivary glands. Blocking actin polymerization with cytochalasin D revealed that Tpm3.1 assembly is dependent on actin assembly. We used time-lapse imaging to determine the timing of the appearance of the actin filament reporter LifeAct-RFP and of Tpm3.1-mNeonGreen on secretory granules in LifeAct-RFP transgenic, Tpm3.1-mNeonGreen and myosin IIA-GFP (GFP-tagged MYH9) knock-in mice. Our findings are consistent with the addition of tropomyosin to actin filaments shortly after the initiation of actin filament nucleation, followed by myosin IIA recruitment.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Tropomiosina/metabolismo , Citoesqueleto de Actina/genética , Actinas/genética , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Cadeias Pesadas de Miosina , Miosina não Muscular Tipo IIA/genética , Ligação Proteica , Vesículas Secretórias/genética , Vesículas Secretórias/metabolismo , Tropomiosina/genética
5.
J Cell Sci ; 130(16): 2696-2706, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28687623

RESUMO

Nonmuscle myosin IIB (NMIIB; heavy chain encoded by MYH10) is essential for cardiac myocyte cytokinesis. The role of NMIIB in other cardiac cells is not known. Here, we show that NMIIB is required in epicardial formation and functions to support myocardial proliferation and coronary vessel development. Ablation of NMIIB in epicardial cells results in disruption of epicardial integrity with a loss of E-cadherin at cell-cell junctions and a focal detachment of epicardial cells from the myocardium. NMIIB-knockout and blebbistatin-treated epicardial explants demonstrate impaired mesenchymal cell maturation during epicardial epithelial-mesenchymal transition. This is manifested by an impaired invasion of collagen gels by the epicardium-derived mesenchymal cells and the reorganization of the cytoskeletal structure. Although there is a marked decrease in the expression of mesenchymal genes, there is no change in Snail (also known as Snai1) or E-cadherin expression. Studies from epicardium-specific NMIIB-knockout mice confirm the importance of NMIIB for epicardial integrity and epicardial functions in promoting cardiac myocyte proliferation and coronary vessel formation during heart development. Our findings provide a novel mechanism linking epicardial formation and epicardial function to the activity of the cytoplasmic motor protein NMIIB.


Assuntos
Diferenciação Celular/genética , Células-Tronco Mesenquimais/fisiologia , Cadeias Pesadas de Miosina/fisiologia , Miosina não Muscular Tipo IIB/fisiologia , Pericárdio/citologia , Pericárdio/embriologia , Animais , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Coração/embriologia , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/genética , Miosina não Muscular Tipo IIB/genética , Organogênese/genética
6.
Dev Biol ; 427(1): 121-130, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28478097

RESUMO

In kidney development, connection of the nephric duct (ND) to the cloaca and subsequent sprouting of the ureteric bud (UB) from the ND are important for urinary exit tract formation. Although the roles of Ret signaling are well established, it remains unclear how intracellular cytoskeletal proteins regulate these morphogenetic processes. Myh9 and Myh10 encode two different non-muscle myosin II heavy chains, and Myh9 mutations in humans are implicated in congenital kidney diseases. Here we report that ND/UB lineage-specific deletion of Myh9/Myh10 in mice caused severe hydroureter/hydronephrosis at birth. At mid-gestation, the mutant ND/UB epithelia exhibited aberrant basal protrusion and ectopic UB formation, which likely led to misconnection of the ureter to the bladder. In addition, the mutant epithelia exhibited apical extrusion followed by massive apoptosis in the lumen, which could be explained by reduced apical constriction and intercellular adhesion mediated by E-cadherin. These phenotypes were not ameliorated by genetic reduction of the tyrosine kinase receptor Ret. In contrast, ERK was activated in the mutant cells and its chemical inhibition partially ameliorated the phenotypes. Thus, myosin II is essential for maintaining the apicobasal integrity of the developing kidney epithelia independently of Ret signaling.


Assuntos
Epitélio/anormalidades , Rim/embriologia , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Ureter/anormalidades , Bexiga Urinária/anormalidades , Animais , Animais Recém-Nascidos , Cães , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Hibridização In Situ , Rim/metabolismo , Células Madin Darby de Rim Canino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Néfrons/anormalidades , Néfrons/metabolismo , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIB/genética , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Ureter/metabolismo , Bexiga Urinária/metabolismo
7.
Traffic ; 15(4): 418-32, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24443954

RESUMO

Variable requirements for actin during clathrin-mediated endocytosis (CME) may be related to regional or cellular differences in membrane tension. To compensate, local regulation of force generation may be needed to facilitate membrane curving and vesicle budding. Force generation is assumed to occur primarily through actin polymerization. Here we examine the role of myosin II using loss of function experiments. Our results indicate that myosin II acts on cortical actin scaffolds primarily in the plane of the plasma membrane (bottom arrow) to generate changes that are critical for enhancing CME progression.


Assuntos
Clatrina/fisiologia , Endocitose/fisiologia , Miosina Tipo II/fisiologia , Actinas/metabolismo , Animais , Camundongos , Camundongos Knockout , Músculos/fisiologia , Miosina Tipo II/genética , Transferrina/metabolismo
8.
Hum Mol Genet ; 23(21): 5706-19, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24908670

RESUMO

Cardiac hypertrophy, an adaptive process that responds to increased wall stress, is characterized by the enlargement of cardiomyocytes and structural remodeling. It is stimulated by various growth signals, of which the mTORC1 pathway is a well-recognized source. Here, we show that loss of Flcn, a novel AMPK-mTOR interacting molecule, causes severe cardiac hypertrophy with deregulated energy homeostasis leading to dilated cardiomyopathy in mice. We found that mTORC1 activity was upregulated in Flcn-deficient hearts, and that rapamycin treatment significantly reduced heart mass and ameliorated cardiac dysfunction. Phospho-AMP-activated protein kinase (AMPK)-alpha (T172) was reduced in Flcn-deficient hearts and nonresponsive to various stimulations including metformin and AICAR (5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide). ATP levels were elevated and mitochondrial function was increased in Flcn-deficient hearts, suggesting that excess energy resulting from up-regulated mitochondrial metabolism under Flcn deficiency might attenuate AMPK activation. Expression of Ppargc1a, a central molecule for mitochondrial metabolism, was increased in Flcn-deficient hearts and indeed, inactivation of Ppargc1a in Flcn-deficient hearts significantly reduced heart mass and prolonged survival. Ppargc1a inactivation restored phospho-AMPK-alpha levels and suppressed mTORC1 activity in Flcn-deficient hearts, suggesting that up-regulated Ppargc1a confers increased mitochondrial metabolism and excess energy, leading to inactivation of AMPK and activation of mTORC1. Rapamycin treatment did not affect the heart size of Flcn/Ppargc1a doubly inactivated hearts, further supporting the idea that Ppargc1a is the critical element leading to deregulation of the AMPK-mTOR-axis and resulting in cardiac hypertrophy under Flcn deficiency. These data support an important role for Flcn in cardiac homeostasis in the murine model.


Assuntos
Cardiomegalia/genética , Cardiomegalia/metabolismo , Estrona/genética , Inativação Gênica , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Trifosfato de Adenosina/biossíntese , Animais , Cardiomegalia/complicações , Cardiomegalia/tratamento farmacológico , Cardiomegalia/patologia , Linhagem Celular , Modelos Animais de Doenças , Ativação Enzimática , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Transgênicos , Renovação Mitocondrial , Tamanho do Órgão/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fosforilação , Transdução de Sinais , Sirolimo/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Função Ventricular/efeitos dos fármacos
9.
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
10.
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
11.
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
12.
J Neurosci ; 33(46): 18149-60, 2013 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-24227724

RESUMO

In the developing brain, cortical GABAergic interneurons migrate long distances from the medial ganglionic eminence (MGE) in which they are generated, to the cortex in which they settle. MGE cells express the cell adhesion molecule N-cadherin, a homophilic cell-cell adhesion molecule that regulates numerous steps of brain development, from neuroepithelium morphogenesis to synapse formation. N-cadherin is also expressed in embryonic territories crossed by MGE cells during their migration. In this study, we demonstrate that N-cadherin is a key player in the long-distance migration of future cortical interneurons. Using N-cadherin-coated substrate, we show that N-cadherin-dependent adhesion promotes the migration of mouse MGE cells in vitro. Conversely, mouse MGE cells electroporated with a construct interfering with cadherin function show reduced cell motility, leading process instability, and impaired polarization associated with abnormal myosin IIB dynamics. In vivo, the capability of electroporated MGE cells to invade the developing cortical plate is altered. Using genetic ablation of N-cadherin in mouse embryos, we show that N-cadherin-depleted MGEs are severely disorganized. MGE cells hardly exit the disorganized proliferative area. N-cadherin ablation at the postmitotic stage, which does not affect MGE morphogenesis, alters MGE cell motility and directionality. The tangential migration to the cortex of N-cadherin ablated MGE cells is delayed, and their radial migration within the cortical plate is perturbed. Altogether, these results identify N-cadherin as a pivotal adhesion substrate that activates cell motility in future cortical interneurons and maintains cell polarity over their long-distance migration to the developing cortex.


Assuntos
Caderinas/metabolismo , Movimento Celular/fisiologia , Polaridade Celular/fisiologia , Córtex Cerebral/metabolismo , Interneurônios/metabolismo , Neurogênese/fisiologia , Animais , Caderinas/deficiência , Células Cultivadas , Córtex Cerebral/citologia , Técnicas de Cocultura , Feminino , Previsões , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Gravidez
13.
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
14.
J Biol Chem ; 288(46): 33398-410, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24072716

RESUMO

Nonmuscle myosin IIs (NM IIs) are a group of molecular motors involved in a wide variety of cellular processes including cytokinesis, migration, and control of cell morphology. There are three paralogs of the NM II heavy chain in humans (IIA, IIB, and IIC), each encoded by a separate gene. These paralogs are expressed at different levels according to cell type and have different roles and intracellular distributions in vivo. Most previous studies on NM II used tissue-purified protein or expressed fragments of the molecule, which presents potential drawbacks for characterizing individual paralogs of the intact protein in vitro. To circumvent current limitations and approach their native properties, we have successfully expressed and purified the three full-length human NM II proteins with their light chains, using the baculovirus/Sf9 system. The enzymatic and structural properties of the three paralogs were characterized. Although each NM II is capable of forming bipolar filaments, those formed by IIC tend to contain fewer constituent molecules than those of IIA and IIB. All paralogs adopt the compact conformation in the presence of ATP. Phosphorylation of the regulatory light chain leads to assembly into filaments, which bind to actin in the presence of ATP. The nature of interactions with actin filaments is shown with different paralogs exhibiting different actin binding behaviors under equivalent conditions. The data show that although NM IIA and IIB form filaments with similar properties, NM IIC forms filaments that are less well suited to roles such as tension maintenance within the cell.


Assuntos
Proteínas Motores Moleculares/química , Proteínas Motores Moleculares/metabolismo , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo II/química , Miosina Tipo II/metabolismo , Miosina não Muscular Tipo IIB/química , Miosina não Muscular Tipo IIB/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Animais , Humanos , Proteínas Motores Moleculares/genética , Cadeias Pesadas de Miosina/genética , Miosina Tipo II/genética , Miosina não Muscular Tipo IIB/genética , Fosforilação/fisiologia , Coelhos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Sf9 , Spodoptera
15.
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
16.
Biochem Biophys Res Commun ; 450(4): 1662-7, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-25044120

RESUMO

Rbfox3, a neuron-specific RNA-binding protein, plays an important role in neuronal differentiation during development. An isoform Rbfox3-d31, which excludes the 93-nucleotide cassette exon within the RNA recognition motif of chicken Rbfox3, has been previously identified. However, the cellular functions of Rbfox3-d31 remain largely unknown. Here we find that Rbfox3-d31 mRNA is highly expressed during the early developmental stages of the chicken embryo, while Rbfox3-d31 protein is barely detected during the same stage due to its rapid degradation mediated by the ubiquitin-proteasome pathway. Importantly, this degradation is specific to the Rbfox3-d31 isoform and it does not occur with full-length Rbfox3. Furthermore, suppression of Rbfox3-d31 protein degradation with the proteasome inhibitor MG132 attenuates the splicing activity of another Rbfox family member Rbfox2 by altering the subcellular localization of Rbfox2. These results suggest that Rbfox3-d31 functions as a repressor for the splicing activity of the Rbfox family and its protein level is regulated in an isoform-specific manner in vivo.


Assuntos
Isoenzimas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Sequência de Bases , Embrião de Galinha , Primers do DNA , Proteínas do Tecido Nervoso/genética , Proteólise , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
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
18.
Nat Cell Biol ; 9(3): 299-309, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17310241

RESUMO

Non-muscle myosin II has diverse functions in cell contractility, cytokinesis and locomotion, but the specific contributions of its different isoforms have yet to be clarified. Here, we report that ablation of the myosin IIA isoform results in pronounced defects in cellular contractility, focal adhesions, actin stress fibre organization and tail retraction. Nevertheless, myosin IIA-deficient cells display substantially increased cell migration and exaggerated membrane ruffling, which was dependent on the small G-protein Rac1, its activator Tiam1 and the microtubule moter kinesin Eg5. Myosin IIA deficiency stabilized microtubules, shifting the balance between actomyosin and microtubules with increased microtubules in active membrane ruffles. When microtubule polymerization was suppressed, myosin IIB could partially compensate for the absence of the IIA isoform in cellular contractility, but not in cell migration. We conclude that myosin IIA negatively regulates cell migration and suggest that it maintains a balance between the actomyosin and microtubule systems by regulating microtubule dynamics.


Assuntos
Actomiosina/metabolismo , Movimento Celular/fisiologia , Microtúbulos/metabolismo , Miosina não Muscular Tipo IIA/fisiologia , Aminoquinolinas/farmacologia , Animais , Azepinas/farmacologia , Células COS , Adesão Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Chlorocebus aethiops , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Cinesinas/antagonistas & inibidores , Cinesinas/genética , Cinesinas/metabolismo , Camundongos , Microtúbulos/efeitos dos fármacos , Naftalenos/farmacologia , Nocodazol/farmacologia , Miosina não Muscular Tipo IIA/antagonistas & inibidores , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIB/antagonistas & inibidores , Miosina não Muscular Tipo IIB/genética , Miosina não Muscular Tipo IIB/fisiologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/genética , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T , Tionas/farmacologia , Transfecção , Vimblastina/farmacologia , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
19.
Dev Biol ; 369(2): 356-61, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22820068

RESUMO

Cytokinesis, the final stage of cell division, bisects the cytoplasm into two daughter cells. In mitotic cells, this process depends on the activity of non-muscle myosin II (NMII), a family of actin-binding motor-proteins that participate in the formation of the cleavage furrow. The relevance of NMII for meiotic cell division, however, is poorly understood. The NMII family consists of three members, NMIIA, NMIIB, and NMIIC, containing different myosin heavy chains (MYH9, MYH10, and MYH14, respectively). We find that a single non-muscle myosin II, NMIIB, is required for meiotic cytokinesis in male but not female mice. Specifically, NMIIB-deficient spermatocytes exhibit cytokinetic failure in meiosis I, resulting in bi-nucleated secondary spermatocytes. Additionally, cytokinetic failure at meiosis II gives rise to bi-nucleated or even tetra-nucleated spermatids. These multi-nucleated spermatids fail to undergo normal differentiation, leading to male infertility. In spite of the presence of multiple non-muscle myosin II isoforms, we demonstrate that a single member, NMIIB, plays an essential and non-redundant role in cytokinesis during meiotic cell divisions of the male germline.


Assuntos
Citocinese/fisiologia , Meiose/fisiologia , Cadeias Pesadas de Miosina/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Espermatogênese/fisiologia , Animais , Divisão Celular/genética , Divisão Celular/fisiologia , Citocinese/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Meiose/genética , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Cadeias Pesadas de Miosina/deficiência , Cadeias Pesadas de Miosina/genética , Miosina não Muscular Tipo IIB/deficiência , Miosina não Muscular Tipo IIB/genética , Espermátides/metabolismo , Espermátides/ultraestrutura , Espermatócitos/metabolismo , Espermatócitos/ultraestrutura , Espermatogênese/genética , Espermatogônias/metabolismo , Espermatogônias/ultraestrutura , Testículo/citologia , Testículo/metabolismo
20.
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
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa