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










Base de dados
Intervalo de ano de publicação
2.
J Cachexia Sarcopenia Muscle ; 9(6): 1109-1120, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30270531

RESUMO

BACKGROUND: Cancer cachexia is a metabolic wasting syndrome that is strongly associated with a poor prognosis. The initiating factors causing fat and muscle loss are largely unknown. Previously, we found that leukaemia inhibitory factor (LIF) secreted by C26 colon carcinoma cells was responsible for atrophy in treated myotubes. In the present study, we tested whether C26 tumour-derived LIF is required for cancer cachexia in mice by knockout of Lif in C26 cells. METHODS: A C26 Lif null tumour cell line was made using CRISPR-Cas9. Measurements of cachexia were compared in mice inoculated with C26 vs. C26Lif-/- tumour cells, and atrophy was compared in myotubes treated with medium from C26 vs. C26Lif-/- tumour cells. Levels of 25 cytokines/chemokines were compared in serum of mice bearing C26 vs. C26Lif-/- tumours and in the medium from these tumour cell lines. RESULTS: At study endpoint, C26 mice showed outward signs of sickness while mice with C26Lif-/- tumours appeared healthy. Mice with C26Lif-/- tumours showed a 55-75% amelioration of body weight loss, muscle loss, fat loss, and splenomegaly compared with mice with C26 tumours (P < 0.05). The heart was not affected by LIF levels because the loss of cardiac mass was the same in C26 and C26Lif-/- tumour-bearing mice. LIF levels in mouse serum was entirely dependent on secretion from the tumour cells. Serum levels of interleukin-6 and G-CSF were increased by 79-fold and 68-fold, respectively, in C26 mice but only by five-fold and two-fold, respectively, in C26Lif-/- mice, suggesting that interleukin-6 and G-CSF increases are dependent on tumour-derived LIF. CONCLUSIONS: This study shows the first use of CRISPR-Cas9 knockout of a candidate cachexia factor in tumour cells. The results provide direct evidence for LIF as a major cachexia initiating factor for the C26 tumour in vivo. Tumour-derived LIF was also a regulator of multiple cytokines in C26 tumour cells and in C26 tumour-bearing mice. The identification of tumour-derived factors such as LIF that initiate the cachectic process is immediately applicable to the development of therapeutics to treat cachexia. This is a proof of principle for studies that when carried out in human cells, will make possible an understanding of the factors causing cachexia in a patient-specific manner.


Assuntos
Caquexia/etiologia , Fator Inibidor de Leucemia/metabolismo , Neoplasias/complicações , Neoplasias/metabolismo , Alelos , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Fator Inibidor de Leucemia/química , Fator Inibidor de Leucemia/genética , Camundongos , Morbidade , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Neoplasias/genética , Tamanho do Órgão , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Front Physiol ; 8: 738, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28993738

RESUMO

Cachexia is strongly associated with a poor prognosis in cancer patients but the biological trigger is unknown and therefore no therapeutics exist. The loss of skeletal muscle is the most deleterious aspect of cachexia and it appears to depend on secretions from tumor cells. Models for studying wasting in cell culture consist of experiments where skeletal muscle cells are incubated with medium conditioned by tumor cells. This has led to candidates for cachectic factors but some of the features of cachexia in vivo are not yet well-modeled in cell culture experiments. Mouse myotube atrophy measured by myotube diameter in response to medium conditioned by mouse colon carcinoma cells (C26) is consistently less than what is seen in muscles of mice bearing C26 tumors with moderate to severe cachexia. One possible reason for this discrepancy is that in vivo the C26 tumor and skeletal muscle share a circulatory system exposing the muscle to tumor factors in a constant and increasing way. We have applied Transwell®-adapted cell culture conditions to more closely simulate conditions found in vivo where muscle is exposed to the ongoing kinetics of constant tumor secretion of active factors. C26 cells were incubated on a microporous membrane (a Transwell® insert) that constitutes the upper compartment of wells containing plated myotubes. In this model, myotubes are exposed to a constant supply of cancer cell secretions in the medium but without direct contact with the cancer cells, analogous to a shared circulation of muscle and cancer cells in tumor-bearing animals. The results for myotube diameter support the idea that the use of Transwell® inserts serves as a more physiological model of the muscle wasting associated with cancer cachexia than the bolus addition of cancer cell conditioned medium. The Transwell® model supports the notion that the dose and kinetics of cachectic factor delivery to muscle play a significant role in the extent of pathology.

4.
J Biol Chem ; 290(32): 19976-86, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26092726

RESUMO

Cachexia is an exacerbating event in many types of cancer that is strongly associated with a poor prognosis. We have identified cytokine, signaling, and transcription factors that are required for cachexia in the mouse C26 colon carcinoma model of cancer. C2C12 myotubes treated with conditioned medium from C26 cancer cells induced atrophy and activated a STAT-dependent reporter gene but not reporter genes dependent on SMAD, FOXO, C/EBP, NF-κB, or AP-1. Of the gp130 family members IL-11, IL-6, oncostatin M (OSM), and leukemia inhibitory factor (LIF), only OSM and LIF were sufficient to activate the STAT reporter in myotubes. LIF was elevated in C26 conditioned medium (CM), but IL-6, OSM, TNFα, and myostatin were not. A LIF-blocking antibody abolished C26 CM-induced STAT reporter activation, STAT3 phosphorylation, and myotube atrophy but blocking antibodies to IL-6 or OSM did not. JAK2 inhibitors also blocked C26 CM-induced STAT reporter activation, STAT3 phosphorylation, and atrophy in myotubes. LIF at levels found in the C26 CM was sufficient for STAT reporter activation and atrophy in myotubes. In vivo, an increase in serum LIF preceded the increase in IL-6 in mice with C26 tumors. Overexpression of a dominant negative Stat3Cß-EGFP gene in myotubes and in mouse muscle blocked the atrophy caused by C26 CM or C26 tumors, respectively. Taken together, these data support an important role of LIF-JAK2-STAT3 in C26 cachexia and point to a therapeutic approach for at least some types of cancer cachexia.


Assuntos
Adenocarcinoma/metabolismo , Caquexia/metabolismo , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica , Janus Quinase 2/metabolismo , Fator Inibidor de Leucemia/metabolismo , Fator de Transcrição STAT3/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Anticorpos Neutralizantes/farmacologia , Caquexia/genética , Caquexia/patologia , Linhagem Celular Tumoral , Tamanho Celular , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Meios de Cultivo Condicionados/farmacologia , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/genética , Fator Inibidor de Leucemia/antagonistas & inibidores , Fator Inibidor de Leucemia/genética , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Oncostatina M/antagonistas & inibidores , Oncostatina M/genética , Oncostatina M/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT3/genética , Transdução de Sinais , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo
5.
BMC Cancer ; 14: 997, 2014 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-25539728

RESUMO

BACKGROUND: Evidence from cachectic cancer patients and animal models of cancer cachexia supports the involvement of Forkhead box O (FoxO) transcription factors in driving cancer-induced skeletal muscle wasting. However, the genome-wide gene networks and associated biological processes regulated by FoxO during cancer cachexia are unknown. We hypothesize that FoxO is a central upstream regulator of diverse gene networks in skeletal muscle during cancer that may act coordinately to promote the wasting phenotype. METHODS: To inhibit endogenous FoxO DNA-binding, we transduced limb and diaphragm muscles of mice with AAV9 containing the cDNA for a dominant negative (d.n.) FoxO protein (or GFP control). The d.n.FoxO construct consists of only the FoxO3a DNA-binding domain that is highly homologous to that of FoxO1 and FoxO4, and which outcompetes and blocks endogenous FoxO DNA binding. Mice were subsequently inoculated with Colon-26 (C26) cells and muscles harvested 26 days later. RESULTS: Blocking FoxO prevented C26-induced muscle fiber atrophy of both locomotor muscles and the diaphragm and significantly spared force deficits. This sparing of muscle size and function was associated with the differential regulation of 543 transcripts (out of 2,093) which changed in response to C26. Bioinformatics analysis of upregulated gene transcripts that required FoxO revealed enrichment of the proteasome, AP-1 and IL-6 pathways, and included several atrophy-related transcription factors, including Stat3, Fos, and Cebpb. FoxO was also necessary for the cancer-induced downregulation of several gene transcripts that were enriched for extracellular matrix and sarcomere protein-encoding genes. We validated these findings in limb muscles and the diaphragm through qRT-PCR, and further demonstrate that FoxO1 and/or FoxO3a are sufficient to increase Stat3, Fos, Cebpb, and the C/EBPß target gene, Ubr2. Analysis of the Cebpb proximal promoter revealed two bona fide FoxO binding elements, which we further establish are necessary for Cebpb promoter activation in response to IL-6, a predominant cytokine in the C26 cancer model. CONCLUSIONS: These findings provide new evidence that FoxO-dependent transcription is a central node controlling diverse gene networks in skeletal muscle during cancer cachexia, and identifies novel candidate genes and networks for further investigation as causative factors in cancer-induced wasting.


Assuntos
Caquexia/etiologia , Neoplasias do Colo/complicações , Neoplasias do Colo/genética , Fatores de Transcrição Forkhead/metabolismo , Redes Reguladoras de Genes , Estudo de Associação Genômica Ampla , Músculo Esquelético/metabolismo , Sequência de Aminoácidos , Animais , Dependovirus/genética , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Vetores Genéticos/genética , Xenoenxertos , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Complexo de Endopeptidases do Proteassoma/metabolismo , Reprodutibilidade dos Testes , Alinhamento de Sequência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transdução Genética
6.
Am J Physiol Cell Physiol ; 306(8): C762-7, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24553183

RESUMO

The muscle-specific ring finger protein 1 (MuRF1) gene is required for most types of skeletal muscle atrophy yet we have little understanding of its transcriptional regulation. The purpose of this study is to identify whether NF-κB and/or FoxO response elements in the MuRF1 promoter are required for MuRF1 gene activation during skeletal muscle atrophy due to the removal of hindlimb weight bearing ("unloading"). Both NF-κB -dependent and FoxO-dependent luciferase reporter activities were significantly increased at 5 days of unloading. Using a 4.4-kb MuRF1 promoter reporter construct, a fourfold increase in reporter (i.e., luciferase) activity was found in rat soleus muscles after 5 days of hindlimb unloading. This activation was abolished by mutagenesis of either of the two distal putative NF-κB sites or all three putative NF-κB sites but not by mutagenesis of all four putative FoxO sites. This work provides the first direct evidence that NF-κB sites, but not FoxO sites, are required for MuRF1 promoter activation in muscle disuse atrophy in vivo.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Proteínas Musculares/metabolismo , Atrofia Muscular/metabolismo , NF-kappa B/metabolismo , Ativação Transcricional/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Feminino , Fatores de Transcrição Forkhead/genética , Elevação dos Membros Posteriores , Proteínas Musculares/genética , NF-kappa B/genética , Regiões Promotoras Genéticas , Ratos , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/genética
7.
PLoS One ; 9(1): e87776, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24489962

RESUMO

Existing data suggest that NF-kappaB signaling is a key regulator of cancer-induced skeletal muscle wasting. However, identification of the components of this signaling pathway and of the NF-κB transcription factors that regulate wasting is far from complete. In muscles of C26 tumor bearing mice, overexpression of dominant negative (d.n.) IKKß blocked muscle wasting by 69% and the IκBα-super repressor blocked wasting by 41%. In contrast, overexpression of d.n. IKKα or d.n. NIK did not block C26-induced wasting. Surprisingly, overexpression of d.n. p65 or d.n. c-Rel did not significantly affect muscle wasting. Genome-wide mRNA expression arrays showed upregulation of many genes previously implicated in muscle atrophy. To test if these upregulated genes were direct targets of NF-κB transcription factors, we compared genome-wide p65 binding to DNA in control and cachectic muscle using ChIP-sequencing. Bioinformatic analysis of ChIP-sequencing data from control and C26 muscles showed very little p65 binding to genes in cachexia and little to suggest that upregulated p65 binding influences the gene expression associated with muscle based cachexia. The p65 ChIP-seq data are consistent with our finding of no significant change in protein binding to an NF-κB oligonucleotide in a gel shift assay, no activation of a NF-κB-dependent reporter, and no effect of d.n.p65 overexpression in muscles of tumor bearing mice. Taken together, these data support the idea that although inhibition of IκBα, and particularly IKKß, blocks cancer-induced wasting, the alternative NF-κB signaling pathway is not required. In addition, the downstream NF-κB transcription factors, p65 and c-Rel do not appear to regulate the transcriptional changes induced by the C26 tumor. These data are consistent with the growing body of literature showing that there are NF-κB-independent substrates of IKKß and IκBα that regulate physiological processes.


Assuntos
Adenocarcinoma/metabolismo , Caquexia/metabolismo , Quinase I-kappa B/metabolismo , Atrofia Muscular/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Masculino , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Transcriptoma
8.
Exp Physiol ; 98(1): 19-24, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22848079

RESUMO

The nuclear factor-κB (NF-κB) signalling pathway is a necessary component of adult skeletal muscle atrophy resulting from systemic illnesses or disuse. Studies showing a role for the NF-κB pathway in muscle disuse include unloading, denervation and immobilization, and studies showing a role for NF-κB in systemic illnesses include cancer, chronic heart failure and acute septic lung injury. Muscle atrophy due to most of these triggers is associated with activation of NF-κB transcriptional activity. With the exception of muscle unloading, however, there is a paucity of data on the NF-κB transcription factors that regulate muscle atrophy, and little is known about which genes are targeted by NF-κB transcription factors during atrophy. Interestingly, in some cases it appears that the amelioration of muscle atrophy by genetic inhibition of NF-κB signalling proteins is due to effects that are independent of the downstream NF-κB transcription factors. These questions are prime areas for investigation if we are to understand a key component of muscle wasting in adult skeletal muscle.


Assuntos
Atrofia Muscular/metabolismo , NF-kappa B/fisiologia , Transdução de Sinais/fisiologia , Adulto , Animais , Regulação da Expressão Gênica , Humanos , Camundongos , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/fisiologia , Ativação Transcricional/fisiologia
9.
Development ; 140(1): 31-42, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23154418

RESUMO

Understanding the molecular mechanisms of skeletal muscle regeneration is crucial to exploiting this pathway for use in tissue repair. Our data demonstrate that the MEF2A transcription factor plays an essential role in skeletal muscle regeneration in adult mice. Injured Mef2a knockout mice display widespread necrosis and impaired myofiber formation. MEF2A controls this process through its direct regulation of the largest known mammalian microRNA (miRNA) cluster, the Gtl2-Dio3 locus. A subset of the Gtl2-Dio3 miRNAs represses secreted Frizzled-related proteins (sFRPs), inhibitors of WNT signaling. Consistent with these data, Gtl2-Dio3-encoded miRNAs are downregulated in regenerating Mef2a knockout muscle, resulting in upregulated sFRP expression and attenuated WNT activity. Furthermore, myogenic differentiation in Mef2a-deficient myoblasts is rescued by overexpression of miR-410 and miR-433, two miRNAs in the Gtl2-Dio3 locus that repress sFRP2, or by treatment with recombinant WNT3A and WNT5A. Thus, miRNA-mediated modulation of WNT signaling by MEF2A is a requisite step for proper muscle regeneration, and represents an attractive pathway for enhancing regeneration of diseased muscle.


Assuntos
Carbocianinas/metabolismo , MicroRNAs/metabolismo , Músculo Esquelético/fisiologia , Fatores de Regulação Miogênica/fisiologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Regeneração/fisiologia , Proteínas Wnt/metabolismo , Animais , Células COS , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Receptores Frizzled/genética , Técnicas de Silenciamento de Genes , Humanos , Fatores de Transcrição MEF2 , Camundongos , Camundongos Knockout , Fatores de Regulação Miogênica/genética , Transdução de Sinais/fisiologia , Regulação para Cima/genética , Proteínas Wnt/fisiologia
10.
PLoS One ; 7(12): e51478, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251550

RESUMO

NF-kappaB transcriptional activation is required for skeletal muscle disuse atrophy. We are continuing to study how the activation of NF-kB regulates the genes that encode the protein products that cause atrophy. Using ChIP-sequencing we found that Bcl-3, an NF-kB transcriptional activator required for atrophy, binds to the promoters of a number of genes whose collective function describes two major aspects of muscle wasting. By means of bioinformatics analysis of ChIP-sequencing data we found Bcl-3 to be directing transcription networks of proteolysis and energy metabolism. The proteolytic arm of the Bcl-3 networks includes many E3 ligases associated with proteasomal protein degradation, including that of the N-end rule pathway. The metabolic arm appears to be involved in organizing the change from oxidative phosphorylation to glycolysis in atrophying muscle. For one gene, MuRF1, ChIP-sequencing data identified the location of Bcl-3 and p50 binding in the promoter region which directed the creation of deletant and base-substitution mutations of MuRF1 promoter constructs to determine the effect on gene transcription. The results provide the first direct confirmation that the NF-kB binding site is involved in the muscle unloading regulation of MuRF1. Finally, we have combined the ChIP-sequencing results with gene expression microarray data from unloaded muscle to map several direct targets of Bcl-3 that are transcription factors whose own targets describe a set of indirect targets for NF-kB in atrophy. ChIP-sequencing provides the first molecular explanation for the finding that Bcl3 knockout mice are resistant to disuse muscle atrophy. Mapping the transcriptional regulation of muscle atrophy requires an unbiased analysis of the whole genome, which we show is now possible with ChIP-sequencing.


Assuntos
Imunoprecipitação da Cromatina , Redes Reguladoras de Genes/genética , Músculo Esquelético/patologia , Atrofia Muscular/genética , Atrofia Muscular/patologia , Proteínas Proto-Oncogênicas/genética , Análise de Sequência de DNA , Fatores de Transcrição/genética , Animais , Proteína 3 do Linfoma de Células B , Feminino , Regulação da Expressão Gênica , Genes Reporter , Loci Gênicos/genética , Genoma , Elevação dos Membros Posteriores , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Anotação de Sequência Molecular , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Atrofia Muscular/fisiopatologia , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Proteólise , Sítio de Iniciação de Transcrição , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/genética , Suporte de Carga
11.
Am J Physiol Cell Physiol ; 303(2): C135-42, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22592403

RESUMO

Muscle atrophy can be triggered by systemic illnesses that are associated with elevated proinflammatory/catabolic cytokines, which, in turn, are thought to contribute to muscle wasting. In this study, we found that the prototypical NF-κB transcription factor, Rel A (p65), is required for NF-κB activation in C2C12 and L6 myotubes due to treatment with exogenous TNF-α, IL-1α, IL-1ß, TNF-related weak inducer of apoptosis, but not IL-6. All five cytokines induced atrophy in C2C12 myotubes, and inhibition of p65 reversed atrophy due to TNF-α, IL-1α, IL-1ß, TNF-related weak inducer of apoptosis, but not IL-6 treatment. p65 was also required for TNF-α-induced increase in atrophy and inflammatory gene expression. TNF-α- and IL-1ß-treated myotubes increased IL-6 protein expression, but use of an IL-6 blocking antibody showed that the IL-6 production did not contribute to atrophy. These data show that p65 is a required transcription factor mediating the catabolic effects of four different cytokines in cultured myotubes, but IL-6 works by a different mechanism.


Assuntos
Citocinas/toxicidade , Fibras Musculares Esqueléticas/patologia , Fator de Transcrição RelA/fisiologia , Animais , Atrofia/etiologia , Atrofia/patologia , Linhagem Celular , Técnicas de Silenciamento de Genes , Interleucina-1alfa/fisiologia , Interleucina-1beta/fisiologia , Interleucina-6/fisiologia , Células L , Camundongos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/fisiologia , Mioblastos/efeitos dos fármacos , Mioblastos/patologia , Mioblastos/fisiologia , Ratos , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/deficiência , Fator de Necrose Tumoral alfa/fisiologia
12.
Methods Mol Biol ; 798: 231-43, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22130840

RESUMO

Plasmid DNA electrotransfer is a direct method of gene delivery to skeletal muscle commonly used to identify endogenous signaling pathways that mediate muscle remodeling or pathological states in adult rodents. When plasmids encoding a protein to be overexpressed are fused to a fluorescent protein or an epitope-tag, plasmid electrotransfer permits visualization of the expressed protein in muscle fibers. Here, we demonstrate the use of electrotransfer of plasmids encoding mutant or wild type proteins to identify the role of the endogenous protein in regulating muscle fiber atrophy. The plasmids used encode a dominant negative form of the inhibitor of kappaB kinase beta (IKKß) fused to green fluorescent protein (GFP), a constitutively active form of IKKα fused to GFP, and a wild type IKKß fused to an HA tag. We show the effects of overexpression of these proteins on rat or mouse fiber size either with disuse atrophy or in normal weight bearing muscle. The effects of overexpressed proteins on myofiber size are assessed by comparing cross-sectional area of the transfected, fluorescent myofibers to the nontransfected, nonfluorescent myofibers. Using optimized intramuscular plasmid DNA injection and electroporation, we illustrate high transfection efficiency with no overt muscle damage using medium sized fusion proteins (105 kDa).


Assuntos
Expressão Gênica , Técnicas de Transferência de Genes , Músculo Esquelético/metabolismo , Plasmídeos/genética , Animais , Eletroporação/métodos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Imuno-Histoquímica , Injeções Intramusculares , Camundongos , Músculo Esquelético/citologia , Plasmídeos/isolamento & purificação , Ratos
13.
FASEB J ; 25(7): 2276-86, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21427212

RESUMO

The calcium-calmodulin-regulated protein phosphatase calcineurin plays an important regulatory role in muscle differentiation, fiber-type determination, hypertrophy, and muscle regeneration. Because calcineurin functions in numerous processes in muscle, multiple mechanisms are likely necessary to ensure that the activity of this phosphatase is appropriately regulated. Here we demonstrate that the muscle-specific scaffolding protein myospryn modulates calcineurin signaling by inhibiting calcineurin-dependent transcriptional activity in C2C12 myoblasts through direct interaction with the enzyme via its noncanonical tripartite motif (TRIM-like). Consistent with these data, transgenic mice overexpressing both the TRIM-like domain of myospryn and constitutively active calcineurin displayed a severe attenuation in the ability of calcineurin to induce a slow-fiber phenotype. Furthermore, transgenic mice overexpressing the TRIM-like domain of myospryn displayed attenuated muscle regeneration after cardiotoxin-induced muscle injury. These results indicate that myospryn functions as a novel inhibitor of the calcineurin signaling pathway in skeletal muscle.


Assuntos
Calcineurina/metabolismo , Proteínas de Transporte/metabolismo , Fibras Musculares de Contração Lenta/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Animais , Animais Recém-Nascidos , Sítios de Ligação , Células COS , Calcineurina/genética , Proteínas de Transporte/genética , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Proteínas Cardiotóxicas de Elapídeos/toxicidade , Crotoxina/toxicidade , Combinação de Medicamentos , Immunoblotting , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Transgênicos , Proteínas Musculares/genética , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiopatologia , Ligação Proteica , Ratos , Regeneração
14.
PLoS One ; 6(1): e16171, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-21249144

RESUMO

Skeletal muscle atrophy is a debilitating condition associated with weakness, fatigue, and reduced functional capacity. Nuclear factor-kappaB (NF-κB) transcription factors play a critical role in atrophy. Knockout of genes encoding p50 or the NF-κB co-transactivator, Bcl-3, abolish disuse atrophy and thus they are NF-κB factors required for disuse atrophy. We do not know however, the genes targeted by NF-κB that produce the atrophied phenotype. Here we identify the genes required to produce disuse atrophy using gene expression profiling in wild type compared to Nfkb1 (gene encodes p50) and Bcl-3 deficient mice. There were 185 and 240 genes upregulated in wild type mice due to unloading, that were not upregulated in Nfkb1⁻/⁻ and Bcl-3⁻/⁻ mice, respectively, and so these genes were considered direct or indirect targets of p50 and Bcl-3. All of the p50 gene targets were contained in the Bcl-3 gene target list. Most genes were involved with protein degradation, signaling, translation, transcription, and transport. To identify direct targets of p50 and Bcl-3 we performed chromatin immunoprecipitation of selected genes previously shown to have roles in atrophy. Trim63 (MuRF1), Fbxo32 (MAFbx), Ubc, Ctsl, Runx1, Tnfrsf12a (Tweak receptor), and Cxcl10 (IP-10) showed increased Bcl-3 binding to κB sites in unloaded muscle and thus were direct targets of Bcl-3. p50 binding to the same sites on these genes either did not change or increased, supporting the idea of p50:Bcl-3 binding complexes. p65 binding to κB sites showed decreased or no binding to these genes with unloading. Fbxo9, Psma6, Psmc4, Psmg4, Foxo3, Ankrd1 (CARP), and Eif4ebp1 did not show changes in p65, p50, or Bcl-3 binding to κB sites, and so were considered indirect targets of p50 and Bcl-3. This work represents the first study to use a global approach to identify genes required to produce the atrophied phenotype with disuse.


Assuntos
Perfilação da Expressão Gênica , Atrofia Muscular/genética , Subunidade p50 de NF-kappa B/genética , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição/genética , Animais , Proteína 3 do Linfoma de Células B , Sítios de Ligação , Camundongos , Camundongos Knockout , Atrofia Muscular/etiologia , Transtornos Musculares Atróficos/genética , Subunidade p50 de NF-kappa B/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , eIF-2 Quinase
15.
Eur J Cancer ; 46(1): 191-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19857958

RESUMO

The mechanisms eliciting cancer cachexia are not well understood. Wasting of skeletal muscle is problematic because it is responsible for the clinical deterioration in cancer patients and for the ability to tolerate cancer treatment. Studies done on animals suggest that nuclear factor of kappa B (NF-kappaB) signalling is important in the progression of muscle wasting due to several types of tumours. However, there are no published studies in humans on the role of NF-kappaB in cancer cachexia. In this project, we studied the rectus abdominis muscle in patients with gastric tumours (n=14) and in age-matched control subjects (n=10) for markers of NF-kappaB activation. Nuclear levels of p65, p50 and Bcl-3 were the same in both groups of subjects. However, phospho-p65 was elevated by 25% in the muscles of cancer patients. In addition, expression of the inhibitor of kappa B alpha (IkappaBalpha) was decreased by 25% in cancer patients. Decreased expression of IkappaBalpha reflects its degradation by one of the IkappaBalpha kinases and is a marker of NF-kappaB activation. Interestingly, there was no correlation between the stage of cancer and the extent of IkappaBalpha decrease, nor was there a correlation between the degree of cachexia and decreased IkappaBalpha levels. This suggests that the activation of NF-kappaB is an early and sustained event in gastric cancer. The work implicates the NF-kappaB signalling in the initiation and progression of cancer cachexia in humans and demonstrates the need for additional study of this pathway; it also recommends NF-kappaB signalling as a therapeutic target for the amelioration of cachexia as has been suggested from studies done on rodents.


Assuntos
Proteínas I-kappa B/metabolismo , Músculo Esquelético/metabolismo , NF-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Gástricas/metabolismo , Idoso , Proteína 3 do Linfoma de Células B , Caquexia/etiologia , Caquexia/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Inibidor de NF-kappaB alfa , Estadiamento de Neoplasias , Proteínas Nucleares/metabolismo , Avaliação Nutricional , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Neoplasias Gástricas/complicações , Neoplasias Gástricas/patologia , Fatores de Transcrição/metabolismo
16.
Exp Cell Res ; 315(19): 3242-9, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19732770

RESUMO

The microtubule cytoskeleton is known to play a role in cell structure and serve as a scaffold for a variety of active molecules in processes as diverse as motility and cell division. The literature on the role of microtubules in signal transduction, however, is marked by inconsistencies. We have investigated a well-studied signaling pathway, TNF-alpha-induced NF-kappaB activation, and found a connection between the stability of microtubules and the regulation of NF-kappaB signaling in C2C12 myotubes. When microtubules are stabilized by paclitaxel (taxol), there is a strong induction of NF-kappaB even in the absence of TNF-alpha . Although there was no additive effect of taxol and TNF-alpha on NF-kappaB activity suggesting a shared mechanism of activation, taxol strongly induced the NF-kappaB reporter in the presence of a TNF receptor (TNFR) blocking antibody while TNF-alpha did not. Both TNF-alpha and taxol induce the degradation of endogenous IkappaBalpha and either taxol or TNF-alpha induction of NF-kappaB activity was blocked by inhibitors of NF-kappaB acting at different sites in the signaling pathway. Both TNF-alpha and taxol strongly induce known NF-kappaB chemokine target genes. On the other hand, if microtubules are destabilized by colchicine, then the induction of NF-kappaB by TNF-alpha or taxol is greatly reduced. Taken together, we surmise that the activity of microtubules is at the level of the TNFR intracellular domain. This phenomenon may indicate a new level of signaling organization in cell biology, actively created by the state of the cytoskeleton, and has ramifications for therapies where microtubule regulating drugs are used.


Assuntos
Microtúbulos/fisiologia , NF-kappa B/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Linhagem Celular , Citoesqueleto , Humanos , Paclitaxel/farmacologia , Receptores do Fator de Necrose Tumoral/metabolismo , Moduladores de Tubulina/farmacologia
17.
FASEB J ; 23(2): 362-70, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18827022

RESUMO

Nuclear factor-kappaB (NF-kappaB) signaling is necessary for many types of muscle atrophy, yet only some of the required components have been identified. Gene transfer of a dominant negative (d.n.) IKKbeta into rat soleus muscles showed complete inhibition of 7-day disuse-induced activation of a kappaB reporter gene, while overexpression of wild-type (w.t.) IKKbeta did not. Overexpression of a d.n. IKKbeta-EGFP fusion protein showed that atrophy was inhibited by 50%, indicating that IKKbeta is required for the atrophy process. Overexpression of constitutively active (c.a.) IKKbeta-EGFP showed a marked increase in NF-kappaB activity and a decrease in fiber size of weight-bearing soleus muscles, while muscles overexpressing w.t. IKKbeta-HA had no effect. The same results were found for IKKalpha; overexpression of a d.n. form of the protein decreased unloading-induced NF-kappaB activation and inhibited atrophy by 50%, while overexpression of the w.t. protein had no effect. Overexpression of a c.a. IKKalpha-EGFP fusion protein showed that IKKalpha was sufficient to activate NF-kappaB activity and induce fiber atrophy in muscle. Overexpression of d.n. IKKbeta plus d.n. IKKalpha showed an additive effect on the inhibition of disuse atrophy (70%), suggesting that both kinases of the IKK complex are required for muscle atrophy. These data show that both IKKalpha and IKKbeta are necessary and sufficient for physiological muscle atrophy.


Assuntos
Quinase I-kappa B/metabolismo , Atrofia Muscular/enzimologia , Animais , Feminino , Genes Reporter/genética , Quinase I-kappa B/genética , Atrofia Muscular/genética , Atrofia Muscular/patologia , NF-kappa B/genética , NF-kappa B/metabolismo , Ratos , Ratos Wistar
18.
Am J Physiol Cell Physiol ; 292(1): C372-82, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16928772

RESUMO

Skeletal muscle atrophy is associated with a marked and sustained activation of nuclear factor-kappaB (NF-kappaB) activity. Previous work showed that p50 is one of the NF-kappaB family members required for this activation and for muscle atrophy. In this work, we tested whether another NF-kappaB family member, c-Rel, is required for atrophy. Because endogenous inhibitory factor kappaBalpha (IkappaBalpha) was activated (i.e., decreased) at 3 and 7 days of muscle disuse (i.e., hindlimb unloading), we also tested if IkappaBalpha, which binds and retains Rel proteins in the cytosol, is required for atrophy and intermediates of the atrophy process. To do this, we electrotransferred a dominant negative IkappaBalpha (IkappaBalphaDeltaN) in soleus muscles, which were either unloaded or weight bearing. IkappaBalphaDeltaN expression abolished the unloading-induced increase in both NF-kappaB activation and total ubiquitinated protein. IkappaBalphaDeltaN inhibited unloading-induced fiber atrophy by 40%. The expression of certain genes known to be upregulated with atrophy were significantly inhibited by IkappaBalphaDeltaN expression during unloading, including MAFbx/atrogin-1, Nedd4, IEX, 4E-BP1, FOXO3a, and cathepsin L, suggesting these genes may be targets of NF-kappaB transcription factors. In contrast, c-Rel was not required for atrophy because the unloading-induced markers of atrophy were the same in c-rel(-/-) and wild-type mice. Thus IkappaBalpha degradation is required for the unloading-induced decrease in fiber size, the increase in protein ubiquitination, activation of NF-kappaB signaling, and the expression of specific atrophy genes, but c-Rel is not. These data represent a significant advance in our understanding of the role of NF-kappaB/IkappaB family members in skeletal muscle atrophy, and they provide new candidate NF-kappaB target genes for further study.


Assuntos
Proteínas I-kappa B/metabolismo , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Proteínas Proto-Oncogênicas c-rel/metabolismo , Animais , Feminino , Corantes Fluorescentes , Genes Dominantes , Genes Reporter , Proteínas de Fluorescência Verde , Proteínas I-kappa B/genética , Camundongos , Camundongos Knockout , Fibras Musculares Esqueléticas/patologia , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Atrofia Muscular/fisiopatologia , Inibidor de NF-kappaB alfa , NF-kappa B/genética , Proteínas Proto-Oncogênicas c-rel/deficiência , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Ubiquitina/metabolismo , Suporte de Carga
19.
FASEB J ; 21(2): 427-37, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17172638

RESUMO

Ubiquitination-dependent proteolysis is a fundamental process underlying skeletal muscle atrophy. Thus, the role of ubiquitin ligases is of great interest. There are no focused studies in muscle on the ubiquitin ligase Nedd4. We first confirmed increased mRNA expression in rat soleus muscles due to 1-14 days of hind limb unloading. Nedd4 protein localized to the sarcolemmal region of muscle fibers. Hind limb unloading, sciatic nerve denervation, starvation, and diabetes led to atrophy of soleus, plantaris, and gastrocnemius muscles, but only unloaded and denervated muscles showed a marked increase in Nedd4 protein expression. This increase was strongly correlated with decreased Notch1 expression, a known target of Nedd4 in other cell types. Overexpression of dominant negative Nedd4 in soleus muscles completely reversed the unloading-induced decrease of Notch1 expression, indicating that Nedd4 is required for Notch1 inactivation. Overexpression of wild-type Nedd4 in soleus muscles of weight bearing rats caused a decrease in Notch1 protein, indicating that Nedd4 is sufficient for Notch1 down-regulation. To further show that Notch1 is a Nedd4 substrate in muscle, conditional overexpression of Nedd4 in C2C12 myotubes induced ubiquitination of Notch1. This is the first finding of a Nedd4 substrate in muscle and of an ubiquitin ligase, the activity of which distinguishes disuse from cachexia atrophy.


Assuntos
Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Receptor Notch1/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Western Blotting , Linhagem Celular , Complexos Endossomais de Distribuição Requeridos para Transporte , Feminino , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Elevação dos Membros Posteriores/fisiologia , Imuno-Histoquímica , Denervação Muscular , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Tono Muscular/fisiologia , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Atrofia Muscular/genética , Atrofia Muscular/fisiopatologia , Mioblastos/citologia , Mioblastos/metabolismo , Ubiquitina-Proteína Ligases Nedd4 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptor Notch1/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcolema/metabolismo , Transfecção , Ubiquitina-Proteína Ligases/genética , Ubiquitinas/metabolismo
20.
Muscle Nerve ; 33(2): 155-65, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16228971

RESUMO

A variety of conditions lead to skeletal muscle atrophy including muscle inactivity or disuse, multiple disease states (i.e., cachexia), fasting, and age-associated atrophy (sarcopenia). Given the impact on mobility in the latter conditions, inactivity could contribute in a secondary manner to muscle atrophy. Because different events initiate atrophy in these different conditions, it seems that the regulation of protein loss may be unique in each case. In fact differences exist between the regulation of the various atrophy conditions, especially sarcopenia, as evidenced in part by comparisons of transcriptional profiles as well as by the unique triggering molecules found in each case. By contrast, recent studies have shown that many of the intracellular signaling molecules and target genes are similar, particularly among the atrophies related to inactivity and cachexia. This review focuses on the most recent findings related to intracellular signaling during muscle atrophy. Key findings are discussed that relate to signaling involving muscle ubiquitin ligases, the IGF/PI3K/Akt pathway, FOXO activity, caspase-3 activity, and NF-kappaB signaling, and an attempt is made to construct a unifying picture of how these data can be connected to better understand atrophy. Once more detailed cellular mechanisms of the atrophy process are understood, more specific interventions can be designed for the attenuation of protein loss.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Músculo Esquelético/fisiopatologia , Atrofia Muscular/fisiopatologia , Transdução de Sinais , Animais , Caspases/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Humanos , Fator de Crescimento Insulin-Like I/fisiologia , Proteínas Musculares/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/patologia , NF-kappa B/fisiologia , Peptídeo Sintases/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Ubiquitina-Proteína Ligases/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...