Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Med ; 20(10): 1182-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25194572

RESUMO

The progressive loss of muscle regenerative capacity with age or disease results in part from a decline in the number and function of satellite cells, the direct cellular contributors to muscle repair. However, little is known about the molecular effectors underlying satellite cell impairment and depletion. Elevated levels of inflammatory cytokines, including interleukin-6 (IL-6), are associated with both age-related and muscle-wasting conditions. The levels of STAT3, a downstream effector of IL-6, are also elevated with muscle wasting, and STAT3 has been implicated in the regulation of self-renewal and stem cell fate in several tissues. Here we show that IL-6-activated Stat3 signaling regulates satellite cell behavior, promoting myogenic lineage progression through myogenic differentiation 1 (Myod1) regulation. Conditional ablation of Stat3 in Pax7-expressing satellite cells resulted in their increased expansion during regeneration, but compromised myogenic differentiation prevented the contribution of these cells to regenerating myofibers. In contrast, transient Stat3 inhibition promoted satellite cell expansion and enhanced tissue repair in both aged and dystrophic muscle. The effects of STAT3 inhibition on cell fate and proliferation were conserved in human myoblasts. The results of this study indicate that pharmacological manipulation of STAT3 activity can be used to counteract the functional exhaustion of satellite cells in pathological conditions, thereby maintaining the endogenous regenerative response and ameliorating muscle-wasting diseases.


Assuntos
Fator de Transcrição STAT3/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Envelhecimento/genética , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Proliferação de Células , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Desenvolvimento Muscular/fisiologia , Proteína MyoD/genética , Proteína MyoD/metabolismo , Fator de Transcrição PAX7/metabolismo , Regeneração/fisiologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Células Satélites de Músculo Esquelético/citologia , Transdução de Sinais
2.
Am J Physiol Endocrinol Metab ; 303(3): E410-21, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22669242

RESUMO

Cachexia, the metabolic dysregulation leading to sustained loss of muscle and adipose tissue, is a devastating complication of cancer and other chronic diseases. Interleukin-6 and related cytokines are associated with muscle wasting in clinical and experimental cachexia, although the mechanisms by which they might induce muscle wasting are unknown. One pathway activated strongly by IL-6 family ligands is the JAK/STAT3 pathway, the function of which has not been evaluated in regulation of skeletal muscle mass. Recently, we showed that skeletal muscle STAT3 phosphorylation, nuclear localization, and target gene expression are activated in C26 cancer cachexia, a model with high IL-6 family ligands. Here, we report that STAT3 activation is a common feature of muscle wasting, activated in muscle by IL-6 in vivo and in vitro and by different types of cancer and sterile sepsis. Moreover, STAT3 activation proved both necessary and sufficient for muscle wasting. In C(2)C(12) myotubes and in mouse muscle, mutant constitutively activated STAT3-induced muscle fiber atrophy and exacerbated wasting in cachexia. Conversely, inhibiting STAT3 pharmacologically with JAK or STAT3 inhibitors or genetically with dominant negative STAT3 and short hairpin STAT3 reduced muscle atrophy downstream of IL-6 or cancer. These results indicate that STAT3 is a primary mediator of muscle wasting in cancer cachexia and other conditions of high IL-6 family signaling. Thus STAT3 could represent a novel therapeutic target for the preservation of skeletal muscle in cachexia.


Assuntos
Caquexia/prevenção & controle , Interleucina-6/metabolismo , Janus Quinases/antagonistas & inibidores , Músculo Esquelético/patologia , Fator de Transcrição STAT3/antagonistas & inibidores , Síndrome de Emaciação/prevenção & controle , Animais , Células CHO , Caquexia/etiologia , Caquexia/genética , Caquexia/patologia , Células Cultivadas , Cricetinae , Cricetulus , Modelos Animais de Doenças , Feminino , Interleucina-6/genética , Interleucina-6/fisiologia , Janus Quinases/genética , Janus Quinases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Proteínas Mutantes/administração & dosagem , Proteínas Mutantes/genética , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Nitrilas , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/administração & dosagem , Pirazóis/farmacologia , Pirimidinas , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Síndrome de Emaciação/genética , Síndrome de Emaciação/metabolismo , Síndrome de Emaciação/patologia
3.
PLoS One ; 6(7): e22538, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21799891

RESUMO

BACKGROUND: Cachexia, or weight loss despite adequate nutrition, significantly impairs quality of life and response to therapy in cancer patients. In cancer patients, skeletal muscle wasting, weight loss and mortality are all positively associated with increased serum cytokines, particularly Interleukin-6 (IL-6), and the presence of the acute phase response. Acute phase proteins, including fibrinogen and serum amyloid A (SAA) are synthesized by hepatocytes in response to IL-6 as part of the innate immune response. To gain insight into the relationships among these observations, we studied mice with moderate and severe Colon-26 (C26)-carcinoma cachexia. METHODOLOGY/PRINCIPAL FINDINGS: Moderate and severe C26 cachexia was associated with high serum IL-6 and IL-6 family cytokines and highly similar patterns of skeletal muscle gene expression. The top canonical pathways up-regulated in both were the complement/coagulation cascade, proteasome, MAPK signaling, and the IL-6 and STAT3 pathways. Cachexia was associated with increased muscle pY705-STAT3 and increased STAT3 localization in myonuclei. STAT3 target genes, including SOCS3 mRNA and acute phase response proteins, were highly induced in cachectic muscle. IL-6 treatment and STAT3 activation both also induced fibrinogen in cultured C2C12 myotubes. Quantitation of muscle versus liver fibrinogen and SAA protein levels indicates that muscle contributes a large fraction of serum acute phase proteins in cancer. CONCLUSIONS/SIGNIFICANCE: These results suggest that the STAT3 transcriptome is a major mechanism for wasting in cancer. Through IL-6/STAT3 activation, skeletal muscle is induced to synthesize acute phase proteins, thus establishing a molecular link between the observations of high IL-6, increased acute phase response proteins and muscle wasting in cancer. These results suggest a mechanism by which STAT3 might causally influence muscle wasting by altering the profile of genes expressed and translated in muscle such that amino acids liberated by increased proteolysis in cachexia are synthesized into acute phase proteins and exported into the blood.


Assuntos
Reação de Fase Aguda/etiologia , Reação de Fase Aguda/metabolismo , Caquexia/complicações , Caquexia/metabolismo , Neoplasias do Colo/complicações , Músculo Esquelético/metabolismo , Fator de Transcrição STAT3/metabolismo , Reação de Fase Aguda/sangue , Reação de Fase Aguda/genética , Animais , Caquexia/sangue , Caquexia/genética , Linhagem Celular Tumoral , Citocinas/sangue , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Fígado/metabolismo , Fígado/patologia , Camundongos , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/patologia , Transcriptoma
4.
Biochem Biophys Res Commun ; 391(3): 1548-54, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20036643

RESUMO

Cachexia, progressive loss of fat and muscle mass despite adequate nutrition, is a devastating complication of cancer associated with poor quality of life and increased mortality. Myostatin is a potent tonic muscle growth inhibitor. We tested how myostatin inhibition might influence cancer cachexia using genetic and pharmacological approaches. First, hypermuscular myostatin null mice were injected with Lewis lung carcinoma or B16F10 melanoma cells. Myostatin null mice were more sensitive to tumor-induced cachexia, losing more absolute mass and proportionately more muscle mass than wild-type mice. Because myostatin null mice lack expression from development, however, we also sought to manipulate myostatin acutely. The histone deacetylase inhibitor Trichostatin A has been shown to increase muscle mass in normal and dystrophic mice by inducing the myostatin inhibitor, follistatin. Although Trichostatin A administration induced muscle growth in normal mice, it failed to preserve muscle in colon-26 cancer cachexia. Finally we sought to inhibit myostatin and related ligands by administration of the Activin receptor extracellular domain/Fc fusion protein, ACVR2B-Fc. Systemic administration of ACVR2B-Fc potently inhibited muscle wasting and protected adipose stores in both colon-26 and Lewis lung carcinoma cachexia, without affecting tumor growth. Enhanced cachexia in myostatin knockouts indicates that host-derived myostatin is not the sole mediator of muscle wasting in cancer. More importantly, skeletal muscle preservation with ACVR2B-Fc establishes that targeting myostatin-family ligands using ACVR2B-Fc or related molecules is an important and potent therapeutic avenue in cancer cachexia.


Assuntos
Receptores de Activinas Tipo II/uso terapêutico , Caquexia/patologia , Músculo Esquelético/efeitos dos fármacos , Distrofias Musculares/prevenção & controle , Miostatina/antagonistas & inibidores , Neoplasias/complicações , Animais , Caquexia/etiologia , Carcinoma Pulmonar de Lewis/complicações , Modelos Animais de Doenças , Folistatina/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Ligantes , Melanoma Experimental/complicações , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Miostatina/genética , Miostatina/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico
5.
Turk J Haematol ; 23(1): 33-6, 2006 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-27265226

RESUMO

Hemophilia A is an X-linked bleeding disorder resulting mostly from heterogeneous point mutations in the factor VIII (F8) gene. Small/large gene deletions, insertions and gross gene rearrangements underlie the molecular pathogenesis of the disease. Two large inversion mutations due to intrachromosomal recombinations between inverted repeats found in intronic sequences and upstream regions of the F8 gene result in severe hemophilia A. The intron 1 inversion mutation is responsible for about 2% of the cases in various populations. Herein, we report the establishment of the long polymerase chain reaction (PCR) intron 1 inversion mutation detection in our laboratory and a similar frequency of 1-2% among Turkish patients.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...