Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 10: 979673, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36340039

RESUMO

Stress-activated mitogen-activated protein kinase kinase 7 (MKK7) is a member of the dual-specificity mitogen-activated protein kinase family. In the human body, MKK7 controls essential physiological processes, including but not limited to proliferation and differentiation in multiple tissues and organs. MKK7, along with the MKK4 pathway, has been implicated in stress-activated activities and biological events that are mediated by c-Jun N-terminal kinase (JNK) signaling. Although numerous studies have been performed to identify the role of JNK in multiple biological processes, there are limited publications that focus on dissecting the independent role of MKK7. Recent research findings have spurred testing via in vivo genetically deficient models, uncovering previously undocumented JNK-independent functions of MKK7. Here we discuss both JNK-dependent and-independent functions of MKK7 in vivo. This review summarizes the role of MKK7 in inflammation, cytokine production, cancer, and neurological diseases.

2.
J Neuromuscul Dis ; 5(1): 59-73, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29278895

RESUMO

BACKGROUND: Mutations in the LAMA2 gene encoding laminin-α2 cause congenital muscular dystrophy Type 1A (MDC1A), a severe recessive disease with no effective treatment. Previous studies have shown that aberrant activation of caspases and cell death through a pathway regulated by BAX and KU70 is a significant contributor to pathogenesis in laminin-α2-deficiency. OBJECTIVES: To identify mechanisms of pathogenesis in MDC1A. METHODS: We used immunocytochemical and molecular studies of human myogenic cells and mouse muscles-comparing laminin-α2-deficient vs. healthy controls-to identify mechanisms that regulate pathological activation of caspase in laminin-α2-deficiency. RESULTS: In cultures of myogenic cells from MDC1A donors, p53 accumulated in a subset of nuclei and aberrant caspase activation was inhibited by the p53 inhibitor pifithrin-alpha. Also, the p53 target BBC3 (PUMA) was upregulated in both MDC1A myogenic cells and Lama2-/- mouse muscles. In addition, studies with sirtuin inhibitors and SIRT1 overexpression showed that caspase activation in MDC1A myotubes was inversely related to sirtuin deacetylase activity. Caspase activation in laminin-α2-deficiency was, however, not associated with increased phosphorylation of p38 MAPK. CONCLUSIONS: Aberrant caspase activation in MDC1A cells was mediated both by sirtuin deacetylase activity and by p53. Interventions that inhibit aberrant caspase activation by targeting sirtuin or p53 function could potentially be useful in ameliorating MDC1A.


Assuntos
Caspases/metabolismo , Laminina/genética , Fibras Musculares Esqueléticas/metabolismo , Distrofias Musculares/metabolismo , Sirtuínas/metabolismo , Células-Tronco/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Benzotiazóis/farmacologia , Histona Desacetilases do Grupo III/metabolismo , Humanos , Laminina/metabolismo , Camundongos , Camundongos Knockout , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/efeitos dos fármacos , Distrofias Musculares/genética , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Sirtuína 1/metabolismo , Células-Tronco/efeitos dos fármacos , Tolueno/análogos & derivados , Tolueno/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Skelet Muscle ; 3(1): 28, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-24314268

RESUMO

BACKGROUND: Congenital muscular dystrophy Type 1A (MDC1A) is a severe, recessive disease of childhood onset that is caused by mutations in the LAMA2 gene encoding laminin-α2. Studies with both mouse models and primary cultures of human MDC1A myogenic cells suggest that aberrant activation of cell death is a significant contributor to pathogenesis in laminin-α2-deficiency. METHODS: To overcome the limited population doublings of primary cultures, we generated immortalized, clonal lines of human MDC1A myogenic cells via overexpression of both CDK4 and the telomerase catalytic component (human telomerase reverse transcriptase (hTERT)). RESULTS: The immortalized MDC1A myogenic cells proliferated indefinitely when cultured at low density in high serum growth medium, but retained the capacity to form multinucleate myotubes and express muscle-specific proteins when switched to low serum medium. When cultured in the absence of laminin, myotubes formed from immortalized MDC1A myoblasts, but not those formed from immortalized healthy or disease control human myoblasts, showed significantly increased activation of caspase-3. This pattern of aberrant caspase-3 activation in the immortalized cultures was similar to that found previously in primary MDC1A cultures and laminin-α2-deficient mice. CONCLUSIONS: Immortalized MDC1A myogenic cells provide a new resource for studies of pathogenetic mechanisms and for screening possible therapeutic approaches in laminin-α2-deficiency.

4.
Biochem Biophys Res Commun ; 362(1): 101-106, 2007 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-17706592

RESUMO

We have assessed whether expressing pancreatic transcription factors Pdx1 and Ngn3 in the liver could induce beta-like cells for curing type I diabetes. When co-expressed in the liver of insulin reporter mice using adenovirus, few cells in the liver were turned into beta-like cells which were marked by lacZ expression by X-gal staining. Insulin promoter assay showed that Pdx1 highly activates insulin promoter when combined with Ngn3. In ex vivo experiments to determine permissiveness of stem/progenitor and fully differentiated cells by ectopic transcription factors, it was found that more islet genes were induced in ES or bone marrow SP cells than fully differentiated NIH3T3 or MEF cells by Pdx1 or Ngn3. Our results suggest that synergistic action of Pdx1 and Ngn3 on inducing various islet genes as well as cell intrinsic factors played a crucial role in directing few liver cells into beta-like cells in vivo.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas de Homeodomínio/fisiologia , Fígado/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Transativadores/fisiologia , Animais , Clonagem Molecular , Diabetes Mellitus/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Transgênicos , Células NIH 3T3 , Regiões Promotoras Genéticas , Ratos , Ativação Transcricional
5.
Mol Ther ; 15(9): 1630-9, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17609656

RESUMO

Given the therapeutic potential offered by embryonic stem (ES) cells, it is critical to optimize stable gene delivery and expression at different developmental stages of ES cell differentiation. Here, we systematically analyzed lentiviral vectors containing the following promoters: the human elongation factor 1alpha (EF1alpha) promoter, the human cytomegalovirus (CMV) immediate early region enhancer-promoter, the composite CAG promoter (consisting of the CMV immediate early enhancer and the chicken beta-actin promoter), the human phosphoglycerate kinase 1 (PGK) promoter, the murine stem cell virus (MSCV) long terminal repeat (LTR), or the gibbon ape leukemia virus (GALV) LTR. Our results show that the EF1alpha promoter directed robust transgene expression at every stage of mouse ES cell differentiation, whereas the CMV promoter drove transgene expression only during late stages. Similarly, the CAG and PGK promoters drove transgene expression at a significant level only during late stages. The MSCV LTR and the GALV LTR exhibited much lower promoter activities at all stages. Interestingly, mouse ES cells transduced with the EF1alpha promoter-containing lentiviral vector lost most of their transgene expression during in vitro differentiation to neural precursors and neuronal cells. Our results demonstrate that different cellular and viral promoters exhibit very distinct and dynamic properties not only in terms of promoter strength but also with respect to differentiation stage-specific activity.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Lentivirus/genética , Regiões Promotoras Genéticas/genética , Animais , Diferenciação Celular/genética , Linhagem Celular , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células-Tronco Embrionárias/citologia , Citometria de Fluxo , Vetores Genéticos/genética , Camundongos , Microscopia de Fluorescência , Fator 1 de Elongação de Peptídeos/genética , Fosfoglicerato Quinase/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
6.
Dev Biol ; 301(1): 70-81, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17150207

RESUMO

We have identified a gene by microarray analysis that is located on chromosome 6 (c6orf32), whose expression is increased during human fetal myoblast differentiation. The protein encoded by c6orf32 is expressed both in myogenic and non-myogenic primary cells isolated from 18-week old human fetal skeletal muscle. Immunofluorescent staining indicated that C6ORF32 localizes to the cellular cytoskeleton and filopodia, and often displays polarized expression within the cell. mRNA knockdown experiments in the C2C12 murine myoblast cell line demonstrated that cells lacking c6orf32 exhibit a myogenic differentiation defect, characterized by a decrease in the expression of myogenin and myosin heavy chain (MHC) proteins, whereas MyoD1 was unaltered. In contrast, overexpression of c6orf32 in C2C12 or HEK293 cells (a non-muscle cell line) promoted formation of long membrane protrusions (filopodia). Analysis of serial deletion mutants demonstrated that amino acids 55-113 of C6ORF32 are likely involved in filopodia formation. These results indicate that C6ORF32 is a novel protein likely to play multiple functions, including promoting myogenic cell differentiation, cytoskeletal rearrangement and filopodia formation.


Assuntos
Diferenciação Celular/fisiologia , Músculos/metabolismo , Proteínas/metabolismo , Regulação para Cima , Sequência de Bases , Moléculas de Adesão Celular , Linhagem Celular , Primers do DNA , Humanos , Músculos/embriologia , Proteínas/genética , RNA Mensageiro/genética
7.
J Cell Sci ; 119(Pt 15): 3117-27, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16835268

RESUMO

Myoblast fusion is a highly regulated process that is important during muscle development and myofiber repair and is also likely to play a key role in the incorporation of donor cells in myofibers for cell-based therapy. Although several proteins involved in muscle cell fusion in Drosophila are known, less information is available on the regulation of this process in vertebrates, including humans. To identify proteins that are regulated during fusion of human myoblasts, microarray studies were performed on samples obtained from human fetal skeletal muscle of seven individuals. Primary muscle cells were isolated, expanded, induced to fuse in vitro, and gene expression comparisons were performed between myoblasts and early or late myotubes. Among the regulated genes, melanoma cell adhesion molecule (M-CAM) was found to be significantly downregulated during human fetal muscle cell fusion. M-CAM expression was confirmed on activated myoblasts, both in vitro and in vivo, and on myoendothelial cells (M-CAM(+) CD31(+)), which were positive for the myogenic markers desmin and MyoD. Lastly, in vitro functional studies using M-CAM RNA knockdown demonstrated that inhibition of M-CAM expression enhances myoblast fusion. These studies identify M-CAM as a novel marker for myogenic progenitors in human fetal muscle and confirm that downregulation of this protein promotes myoblast fusion.


Assuntos
Biomarcadores/metabolismo , Antígeno CD146/metabolismo , Fusão Celular , Feto/anatomia & histologia , Músculo Esquelético , Mioblastos/fisiologia , Adulto , Animais , Antígeno CD146/genética , Fracionamento Celular , Células Cultivadas , Células Endoteliais/metabolismo , Feminino , Perfilação da Expressão Gênica , Idade Gestacional , Humanos , Músculo Esquelético/citologia , Músculo Esquelético/embriologia , Músculo Esquelético/fisiologia , Mioblastos/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Interferência de RNA
8.
Science ; 298(5593): 597-600, 2002 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-12228720

RESUMO

The transcriptional profiles of mouse embryonic, neural, and hematopoietic stem cells were compared to define a genetic program for stem cells. A total of 216 genes are enriched in all three types of stem cells, and several of these genes are clustered in the genome. When compared to differentiated cell types, stem cells express a significantly higher number of genes (represented by expressed sequence tags) whose functions are unknown. Embryonic and neural stem cells have many similarities at the transcriptional level. These results provide a foundation for a more detailed understanding of stem cell biology.


Assuntos
Embrião de Mamíferos/citologia , Perfilação da Expressão Gênica , Expressão Gênica , Células-Tronco Hematopoéticas/fisiologia , Neurônios/citologia , Células-Tronco/fisiologia , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Células da Medula Óssea/fisiologia , Diferenciação Celular , DNA Helicases/genética , DNA Helicases/metabolismo , Etiquetas de Sequências Expressas , Regulação da Expressão Gênica , Ventrículos Laterais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo , Reprodutibilidade dos Testes , Transdução de Sinais , Transcrição Gênica
9.
J Gen Virol ; 81(Pt 9): 2189-2194, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10950976

RESUMO

S2 is an accessory protein of equine infectious anaemia virus (EIAV), the function of which is unknown. In order to gain insight into the function of S2, the intracellular localization of the protein, its interaction with viral proteins and its incorporation into viral particles have been investigated. Immunolocalization of S2 revealed punctate staining in the cytoplasm and the S2 protein co-precipitated with the EIAV Gag precursor. Despite overexpression of S2 through the use of a codon-optimized sequence, there was no preferential association of S2 with EIAV particles. These data suggest that S2 may function to organize the Gag protein during particle assembly in the cytoplasm but that it is unlikely to be involved in the early stages of the virus life-cycle.


Assuntos
Proteínas Virais/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Humanos , Dados de Sequência Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA