Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Cell Res ; 433(2): 113820, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-37879549

RESUMO

The Warburg effect links growth and glycolysis in cancer. A key purpose of the Warburg effect is to generate glycolytic intermediates for anabolic reactions, such as nucleotides → RNA/DNA and amino acids → protein synthesis. The aim of this study was to investigate whether a similar 'glycolysis-for-anabolism' metabolic reprogramming also occurs in hypertrophying skeletal muscle. To interrogate this, we first induced C2C12 myotube hypertrophy with IGF-1. We then added 14C glucose to the differentiation medium and measured radioactivity in isolated protein and RNA to establish whether 14C had entered anabolism. We found that especially protein became radioactive, suggesting a glucose → glycolytic intermediates → non-essential amino acid(s) → protein series of reactions, the rate of which was increased by IGF-1. Next, to investigate the importance of glycolytic flux and non-essential amino acid synthesis for myotube hypertrophy, we exposed C2C12 and primary mouse myotubes to the glycolysis inhibitor 2-Deoxy-d-glucose (2DG). We found that inhibiting glycolysis lowered C2C12 and primary myotube size. Similarly, siRNA silencing of PHGDH, the key enzyme of the serine biosynthesis pathway, decreased C2C12 and primary myotube size; whereas retroviral PHGDH overexpression increased C2C12 myotube size. Together these results suggest that glycolysis is important for hypertrophying myotubes, which reprogram their metabolism to facilitate anabolism, similar to cancer cells.


Assuntos
Fator de Crescimento Insulin-Like I , Neoplasias , Animais , Camundongos , Fator de Crescimento Insulin-Like I/metabolismo , Fosfoglicerato Desidrogenase/genética , Fosfoglicerato Desidrogenase/metabolismo , Fosfoglicerato Desidrogenase/farmacologia , Fibras Musculares Esqueléticas/metabolismo , Neoplasias/metabolismo , RNA/metabolismo , Hipertrofia/metabolismo , Glucose/farmacologia , Aminoácidos/genética , Aminoácidos/metabolismo , Aminoácidos/farmacologia
2.
Elife ; 112022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35323108

RESUMO

In skeletal muscle, transforming growth factor-ß (TGF-ß) family growth factors, TGF-ß1 and myostatin, are involved in atrophy and muscle wasting disorders. Simultaneous interference with their signalling pathways may improve muscle function; however, little is known about their individual and combined receptor signalling. Here, we show that inhibition of TGF-ß signalling by simultaneous muscle-specific knockout of TGF-ß type I receptors Tgfbr1 and Acvr1b in mice, induces substantial hypertrophy, while such effect does not occur by single receptor knockout. Hypertrophy is induced by increased phosphorylation of Akt and p70S6K and reduced E3 ligases expression, while myonuclear number remains unaltered. Combined knockout of both TGF-ß type I receptors increases the number of satellite cells, macrophages and improves regeneration post cardiotoxin-induced injury by stimulating myogenic differentiation. Extra cellular matrix gene expression is exclusively elevated in muscle with combined receptor knockout. Tgfbr1 and Acvr1b are synergistically involved in regulation of myofibre size, regeneration, and collagen deposition.


Assuntos
Desenvolvimento Muscular , Músculo Esquelético , Animais , Hipertrofia , Camundongos , Músculo Esquelético/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Fator de Crescimento Transformador beta/metabolismo
3.
Aging (Albany NY) ; 14(1): 28-53, 2022 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-35023852

RESUMO

Aging-associated muscle wasting and impaired regeneration are caused by deficiencies in muscle stem cell (MuSC) number and function. We postulated that aged MuSCs are intrinsically impaired in their responsiveness to omnipresent mechanical cues through alterations in MuSC morphology, mechanical properties, and number of integrins, culminating in impaired proliferative capacity. Here we show that aged MuSCs exhibited significantly lower growth rate and reduced integrin-α7 expression as well as lower number of phospho-paxillin clusters than young MuSCs. Moreover, aged MuSCs were less firmly attached to matrigel-coated glass substrates compared to young MuSCs, as 43% of the cells detached in response to pulsating fluid shear stress (1 Pa). YAP nuclear localization was 59% higher than in young MuSCs, yet YAP target genes Cyr61 and Ctgf were substantially downregulated. When subjected to pulsating fluid shear stress, aged MuSCs exhibited reduced upregulation of proliferation-related genes. Together these results indicate that aged MuSCs exhibit impaired mechanosensitivity and growth potential, accompanied by altered morphology and mechanical properties as well as reduced integrin-α7 expression. Aging-associated impaired muscle regenerative capacity and muscle wasting is likely due to aging-induced intrinsic MuSC alterations and dysfunctional mechanosensitivity.


Assuntos
Proliferação de Células/fisiologia , Senescência Celular/fisiologia , Mecanotransdução Celular/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Células-Tronco/fisiologia , Envelhecimento , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Adesão Celular/fisiologia , Cadeias alfa de Integrinas/genética , Cadeias alfa de Integrinas/metabolismo , Camundongos , Óxido Nítrico/metabolismo , Resistência ao Cisalhamento
4.
Cells ; 9(2)2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32041253

RESUMO

Transforming Growth Factor ß (TGF-ß) is involved in fibrosis as well as the regulation of muscle mass, and contributes to the progressive pathology of muscle wasting disorders. However, little is known regarding the time-dependent signalling of TGF-ß in myoblasts and myotubes, as well as how TGF-ß affects collagen type I expression and the phenotypes of these cells. Here, we assessed effects of TGF-ß on gene expression in C2C12 myoblasts and myotubes after 1, 3, 9, 24 and 48 h treatment. In myoblasts, various myogenic genes were repressed after 9, 24 and 48 h, while in myotubes only a reduction in Myh3 expression was observed. In both myoblasts and myotubes, TGF-ß acutely induced the expression of a subset of genes involved in fibrosis, such as Ctgf and Fgf-2, which was subsequently followed by increased expression of Col1a1. Knockdown of Ctgf and Fgf-2 resulted in a lower Col1a1 expression level. Furthermore, the effects of TGF-ß on myogenic and fibrotic gene expression were more pronounced than those of myostatin, and knockdown of TGF-ß type I receptor Tgfbr1, but not receptor Acvr1b, resulted in a reduction in Ctgf and Col1a1 expression. These results indicate that, during muscle regeneration, TGF-ß induces fibrosis via Tgfbr1 by stimulating the autocrine signalling of Ctgf and Fgf-2.


Assuntos
Colágeno Tipo I/metabolismo , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Células Cultivadas , Fibrose , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Desenvolvimento Muscular/efeitos dos fármacos , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/efeitos dos fármacos , Mioblastos/efeitos dos fármacos , Miostatina/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Fatores de Tempo
5.
Adv Exp Med Biol ; 1088: 109-137, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30390250

RESUMO

Insulin-like growth factor 1 (IGF-1) is a key anabolic growth factor stimulating phosphatidylinositol 3-kinase (PI3K)/Akt signaling which is well known for regulating muscle hypertrophy. However, the role of IGF-1 in muscle atrophy is less clear. This review provides an overview of the mechanisms via which IGF-1 signaling is implicated in several conditions of muscle atrophy and via which mechanisms protein turnover is altered. IGF-1/PI3K/Akt signaling stimulates the rate of protein synthesis via p70S6Kinase and p90 ribosomal S6 kinase and negatively regulates protein degradation, predominantly by its inhibiting effect on proteasomal and lysosomal protein degradation. Caspase-dependent protein degradation is also attenuated by IGF/PI3K/Akt signaling, whereas evidence for an effect on calpain-dependent protein degradation is inconclusive. IGF-1/PI3K/Akt signaling reduces during denervation-, unloading-, and joint immobilization-induced muscle atrophy, whereas IGF-1/PI3K/Akt signaling seems unaltered during aging-associated muscle atrophy. During denervation and aging, IGF-1 overexpression or injection counteracts denervation- and aging-associated muscle atrophy, despite enhanced anabolic resistance with regard to IGF-1 signaling with aging. It remains unclear whether pharmacological stimulation of IGF-1/PI3K/Akt signaling attenuates immobilization- or unloading-induced muscle atrophy. Exploration of the possibilities to interfere with IGF-1/PI3K/Akt signaling reveals that microRNAs targeting IGF-1 signaling components are promising targets to counterbalance muscle atrophy. Overall, the findings summarized in this review show that in disuse conditions, but not with aging, IGF-1/PI3K/Akt signaling is attenuated and that in some conditions stimulation of this pathway may alleviate skeletal muscle atrophy.


Assuntos
Fator de Crescimento Insulin-Like I/metabolismo , Atrofia Muscular/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Humanos , Músculo Esquelético/fisiopatologia , Atrofia Muscular/fisiopatologia
6.
Adv Exp Med Biol ; 1088: 153-206, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30390252

RESUMO

Myostatin was identified more than 20 years ago as a negative regulator of muscle mass in mice and cattle. Since then, a wealth of studies have uncovered the potential involvement of myostatin in muscle atrophy and sparked interest in myostatin as a promising therapeutic target to counteract decline of muscle mass in patients afflicted with different muscle-wasting conditions. Insight in the molecular mechanism of myostatin signaling and regulation of myostatin activity has resulted in the identification of specific treatments to inhibit myostatin signaling and related signaling pathways. Currently, several treatments that target myostatin and related proteins have been evaluated in preclinical animal models of muscle wasting, and some potential therapies have progressed to clinical trials. However, studies also revealed potential downsides of myostatin targeting in skeletal muscle and other tissues, which raises the question if myostatin is indeed a valuable target to counteract muscle atrophy. In this review we provide an updated overview of the molecular mechanisms of myostatin signaling, the preclinical evidence supporting a role for myostatin and related proteins in muscle atrophy, and the potential issues that arise when targeting myostatin. In addition, we evaluate the current clinical status of different treatments aimed at inhibiting myostatin and discuss future perspectives of targeting myostatin to counteract muscle atrophy.


Assuntos
Terapia de Alvo Molecular , Músculo Esquelético/patologia , Atrofia Muscular/tratamento farmacológico , Miostatina/antagonistas & inibidores , Transdução de Sinais , Animais , Bovinos , Humanos , Camundongos , Atrofia Muscular/fisiopatologia
7.
J Am Soc Nephrol ; 27(12): 3589-3599, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27020852

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD), characterized by the formation of numerous kidney cysts, is caused by PKD1 or PKD2 mutations and affects 0.1% of the population. Although recent clinical studies indicate that reduction of cAMP levels slows progression of PKD, this finding has not led to an established safe and effective therapy for patients, indicating the need to find new therapeutic targets. The role of TGF-ß in PKD is not clearly understood, but nuclear accumulation of phosphorylated SMAD2/3 in cyst-lining cells suggests the involvement of TGF-ß signaling in this disease. In this study, we ablated the TGF-ß type 1 receptor (also termed activin receptor-like kinase 5) in renal epithelial cells of PKD mice, which had little to no effect on the expression of SMAD2/3 target genes or the progression of PKD. Therefore, we investigated whether alternative TGF-ß superfamily ligands account for SMAD2/3 activation in cystic epithelial cells. Activins are members of the TGF-ß superfamily and drive SMAD2/3 phosphorylation via activin receptors, but activins have not been studied in the context of PKD. Mice with PKD had increased expression of activin ligands, even at early stages of disease. In addition, treatment with a soluble activin receptor IIB fusion (sActRIIB-Fc) protein, which acts as a soluble trap to sequester activin ligands, effectively inhibited cyst formation in three distinct mouse models of PKD. These data point to activin signaling as a key pathway in PKD and a promising target for therapy.


Assuntos
Ativinas/antagonistas & inibidores , Doenças Renais Policísticas/prevenção & controle , Transdução de Sinais , Animais , Progressão da Doença , Células Epiteliais , Feminino , Rim/citologia , Masculino , Camundongos , Doenças Renais Policísticas/etiologia , Proteínas Recombinantes de Fusão/farmacologia , Proteína Smad2/fisiologia , Proteína Smad3/fisiologia , Fatores de Tempo
8.
PLoS One ; 8(7): e69096, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23861958

RESUMO

Fibrodysplasia ossificans progressiva (FOP) is a rare heritable disease characterized by progressive heterotopic ossification of connective tissues, for which there is presently no definite treatment. A recurrent activating mutation (c.617G→A; R206H) of activin receptor-like kinase 2 (ACVR1/ALK2), a BMP type I receptor, has been shown as the main cause of FOP. This mutation constitutively activates the BMP signaling pathway and initiates the formation of heterotopic bone. In this study, we have designed antisense oligonucleotides (AONs) to knockdown mouse ALK2 expression by means of exon skipping. The ALK2 AON could induce exon skipping in cells, which was accompanied by decreased ALK2 mRNA levels and impaired BMP signaling. In addition, the ALK2 AON potentiated muscle differentiation and repressed BMP6-induced osteoblast differentiation. Our results therefore provide a potential therapeutic approach for the treatment of FOP disease by reducing the excessive ALK2 activity in FOP patients.


Assuntos
Receptores de Ativinas Tipo I/antagonistas & inibidores , Éxons , Células Musculares/citologia , Mioblastos/citologia , Miosite Ossificante/terapia , Oligonucleotídeos Antissenso/genética , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Animais , Proteína Morfogenética Óssea 6/genética , Proteína Morfogenética Óssea 6/metabolismo , Diferenciação Celular , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Terapia Genética/métodos , Humanos , Camundongos , Células Musculares/metabolismo , Mutação , Mioblastos/metabolismo , Miosite Ossificante/genética , Miosite Ossificante/metabolismo , Miosite Ossificante/patologia , Oligonucleotídeos Antissenso/síntese química , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteogênese/genética , Transdução de Sinais
9.
Am J Physiol Endocrinol Metab ; 305(2): E171-82, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23695214

RESUMO

The importance of adequate levels of muscle size and function and physical activity is widely recognized. Myostatin/activin blocking increases skeletal muscle mass but may decrease muscle oxidative capacity and can thus be hypothesized to affect voluntary physical activity. Soluble activin receptor IIB (sActRIIB-Fc) was produced to block myostatin/activins. Modestly dystrophic mdx mice were injected with sActRIIB-Fc or PBS with or without voluntary wheel running exercise for 7 wk. Healthy mice served as controls. Running for 7 wk attenuated the sActRIIB-Fc-induced increase in body mass by decreasing fat mass. Running also enhanced/restored the markers of muscle oxidative capacity and autophagy in mdx mice to or above the levels of healthy mice. Voluntary running activity was decreased by sActRIIB-Fc during the first 3-4 wk correlating with increased body mass. Home cage physical activity of mice, quantified from the force plate signal, was decreased by sActRIIB-Fc the whole 7-wk treatment in sedentary mice. To understand what happens during the first weeks after sActRIIB-Fc administration, when mice are less active, healthy mice were injected with sActRIIB-Fc or PBS for 2 wk. During the sActRIIB-Fc-induced rapid 2-wk muscle growth period, oxidative capacity and autophagy were reduced, which may possibly explain the decreased running activity. These results show that increased muscle size and decreased markers of oxidative capacity and autophagy during the first weeks of myostatin/activin blocking are associated with decreased voluntary activity levels. Voluntary exercise in dystrophic mice enhances the markers of oxidative capacity and autophagy to or above the levels of healthy mice.


Assuntos
Receptores de Activinas Tipo II/farmacologia , Ativinas/antagonistas & inibidores , Autofagia/fisiologia , Atividade Motora/fisiologia , Miostatina/antagonistas & inibidores , Condicionamento Físico Animal/fisiologia , Receptores de Activinas Tipo II/biossíntese , Ativinas/fisiologia , Adiposidade/genética , Adiposidade/fisiologia , Animais , Western Blotting , Peso Corporal/fisiologia , Citrato (si)-Sintase/metabolismo , Creatina Quinase/sangue , DNA/biossíntese , DNA/isolamento & purificação , Ingestão de Alimentos/fisiologia , Hematócrito , Hemoglobinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Miostatina/fisiologia , Oxirredução , Fator de Necrose Tumoral alfa/metabolismo
10.
Am J Physiol Endocrinol Metab ; 304(1): E41-50, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23115080

RESUMO

Loss of muscle mass and function occurs in various diseases. Myostatin blocking can attenuate muscle loss, but downstream signaling is not well known. Therefore, to elucidate associated signaling pathways, we used the soluble activin receptor IIb (sActRIIB-Fc) to block myostatin and activins in mice. Within 2 wk, the treatment rapidly increased muscle size as expected but decreased capillary density per area. sActRIIB-Fc increased muscle protein synthesis 1-2 days after the treatment correlating with enhanced mTORC1 signaling (phosphorylated rpS6 and S6K1, r = 0.8). Concurrently, increased REDD1 and eIF2Bε protein contents and phosphorylation of 4E-BP1 and AMPK was observed. In contrast, proangiogenic MAPK signaling and VEGF-A protein decreased. Hippo signaling has been characterized recently as a regulator of organ size and an important regulator of myogenesis in vitro. The phosphorylation of YAP (Yes-associated protein), a readout of activated Hippo signaling, increased after short- and longer-term myostatin and activin blocking and in exercised muscle. Moreover, dystrophic mdx mice had elevated phosphorylated and especially total YAP protein content. These results show that the blocking of myostatin and activins induce rapid skeletal muscle growth. This is associated with increased protein synthesis and mTORC1 signaling but decreased capillary density and proangiogenic signaling. It is also shown for the first time that Hippo signaling is activated in skeletal muscle after myostatin blocking and exercise and also in dystrophic muscle. This suggests that Hippo signaling may have a role in skeletal muscle in various circumstances.


Assuntos
Capilares/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Complexos Multiproteicos/fisiologia , Proteínas Musculares/biossíntese , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Ativinas/antagonistas & inibidores , Animais , Capilares/citologia , Contagem de Células , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Via de Sinalização Hippo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Miostatina/antagonistas & inibidores , Biossíntese de Proteínas/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
11.
Cell Mol Life Sci ; 70(3): 407-23, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22752156

RESUMO

Bone morphogenetic proteins (BMPs) are important extracellular cytokines that play critical roles in embryogenesis and tissue homeostasis. BMPs signal via transmembrane type I and type II serine/threonine kinase receptors and intracellular Smad effector proteins. BMP signaling is precisely regulated and perturbation of BMP signaling is connected to multiple diseases, including musculoskeletal diseases. In this review, we will summarize the recent progress in elucidation of BMP signal transduction, how overactive BMP signaling is involved in the pathogenesis of heterotopic ossification and Duchenne muscular dystrophy, and discuss possible therapeutic strategies for treatment of these diseases.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Ossificação Heterotópica/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Humanos , Distrofia Muscular de Duchenne/patologia , Ossificação Heterotópica/patologia , Transdução de Sinais
12.
Hum Gene Ther ; 23(12): 1269-79, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22894762

RESUMO

Adeno-associated virus (AAV)-U7-mediated skipping of dystrophin-exon-23 restores dystrophin expression and muscle function in the mdx mouse model of Duchenne muscular dystrophy. Soluble activin receptor IIB (sActRIIB-Fc) inhibits signaling of myostatin and homologous molecules and increases muscle mass and function of wild-type and mdx mice. We hypothesized that combined treatment with AAV-U7 and sActRIIB-Fc may synergistically improve mdx muscle function. Bioactivity of sActRIIB-Fc on skeletal muscle was first demonstrated in wild-type mice. In mdx mice we show that AAV-U7-mediated dystrophin restoration improved specific muscle force and resistance to eccentric contractions when applied alone. Treatment of mdx mice with sActRIIB-Fc increased body weight, muscle mass and myofiber size, but had little effect on muscle function. Combined treatment stimulated muscle growth comparable to the effect of sActRIIB-Fc alone and dystrophin rescue was similar to AAV-U7 alone. Moreover, combined treatment improved maximal tetanic force and the resistance to eccentric contraction to similar extent as AAV-U7 alone. In conclusion, combination of dystrophin exon skipping with sActRIIB-Fc brings together benefits of each treatment; however, we failed to evidence a clear synergistic effect on mdx muscle function.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Dependovirus/genética , Distrofina/genética , Terapia Genética/métodos , Distrofia Muscular Animal/terapia , Receptores de Activinas Tipo II/genética , Animais , Peso Corporal , Distrofina/metabolismo , Éxons , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Contração Muscular/genética , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia
13.
Curr Gene Ther ; 12(3): 245-59, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22554312

RESUMO

Since the discovery of the myostatin/ActRIIB signaling pathway 15 years ago, numerous strategies were developed to block its inhibitory function during skeletal muscle growth. Accumulating evidence demonstrates that abrogation of myostatin/ActRIIB signaling ameliorates pathology and function of dystrophic muscle in animal models for Duchenne muscular dystrophy (DMD). Therapeutic trials in healthy man and muscular dystrophy patients suggest feasibility of blockade strategies for potential clinical use. However, many key questions on the effect of myostatin/ActRIIB blockade remain unresolved; such as the underlying molecular mechanism that triggers muscle growth, the effect on muscle regeneration and adult muscle stem cell regulation and whether it causes long term metabolic alterations. Current therapeutic strategies aim to systemically abrogate myostatin/ActRIIB signaling. Although this ensures widespread effect on musculature, it also interferes with ActRIIB signaling in other tissues than skeletal muscle, thereby risking adverse effects. This review discusses current knowledge on myostatin/ActRIIB signaling and its potential value as a therapeutic target for DMD.


Assuntos
Receptores de Activinas Tipo II , Terapia de Alvo Molecular , Distrofia Muscular de Duchenne , Miostatina/antagonistas & inibidores , Receptores de Activinas Tipo II/antagonistas & inibidores , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animais , Ensaios Clínicos Fase II como Assunto , Humanos , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Miostatina/genética , Miostatina/metabolismo , Transdução de Sinais
14.
Cardiovasc Res ; 94(3): 439-49, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22419669

RESUMO

AIM: Treatment of disorders of the sinus node or the atrioventricular node requires insights into the molecular mechanisms of development and homoeostasis of these pacemaker tissues. In the developing heart, transcription factor TBX3 is required for pacemaker and conduction system development. Here, we explore the role of TBX3 in the adult heart and investigate whether TBX3 is able to reprogramme terminally differentiated working cardiomyocytes into pacemaker cells. METHODS AND RESULTS: TBX3 expression was ectopically induced in cardiomyocytes of adult transgenic mice using tamoxifen. Expression analysis revealed an efficient switch from the working myocardial expression profile to that of the pacemaker myocardium. This included suppression of genes encoding gap junction subunits (Cx40, Cx43), the cardiac Na(+) channel (Na(V)1.5; I(Na)), and inwardly rectifying K(+) ion channels (K(ir) genes; I(K1)). Concordantly, we observed conduction slowing in these hearts and reductions in I(Na) and I(K1) in cardiomyocytes isolated from these hearts. The reduction in I(K1) resulted in a more depolarized maximum diastolic potential, thus enabling spontaneous diastolic depolarization. Neither ectopic pacemaker activity nor pacemaker current I(f) was observed. Lentiviral expression of TBX3 in ventricular cardiomyocytes resulted in conduction slowing and development of heterogeneous phenotypes, including depolarized and spontaneously active cardiomyocytes. CONCLUSIONS: TBX3 reprogrammes terminally differentiated working cardiomyocytes and induces important pacemaker properties. The ability of TBX3 to reduce intercellular coupling to overcome current-to-load mismatch and the ability to reduce I(K1) density to enable diastolic depolarization are promising TBX3 characteristics that may facilitate biological pacemaker formation strategies.


Assuntos
Relógios Biológicos/genética , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Miócitos Cardíacos/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Nó Sinoatrial/metabolismo , Proteínas com Domínio T/genética
15.
Neuromuscul Disord ; 22(5): 406-17, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22284942

RESUMO

The genetic defect of mdx mice resembles that of Duchenne muscular dystrophy, although their functional performance and life expectancy is nearly normal. By contrast, mice lacking utrophin and dystrophin (mdx/utrn -/-) are severely affected and die prematurely. Mice with one utrophin allele (mdx/utrn +/-) are more severely affected than mdx mice, but outlive mdx/utrn -/- mice. We subjected mdx/utrn +/+, +/-, -/- and wild type males to a 12week functional test regime of four different functional tests. Mdx/utrn +/+ and +/- mice completed the regime, while mdx/utrn -/- mice died prematurely. Mdx/utrn +/- mice performed significantly worse compared to mdx/utrn +/+ mice in functional tests. Creatine kinase levels, percentage of fibrotic/necrotic tissue, morphology of neuromuscular synapses and expression of biomarker genes were comparable, whereas mdx/utrn +/- and -/- mice had increased levels of regenerating fibers. This makes mdx/utrn +/- mice valuable for testing the benefit of potential therapies on muscle function parameters.


Assuntos
Distrofina/metabolismo , Atividade Motora , Músculo Esquelético/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Utrofina/metabolismo , Animais , Modelos Animais de Doenças , Distrofina/deficiência , Camundongos , Camundongos Endogâmicos mdx , Camundongos Knockout , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/patologia , Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/fisiopatologia , Análise de Sobrevida , Utrofina/deficiência
16.
BMC Med Genomics ; 4: 36, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21507246

RESUMO

BACKGROUND: Myostatin is a potent muscle growth inhibitor that belongs to the Transforming Growth Factor-ß (TGF-ß) family. Mutations leading to non functional myostatin have been associated with hypermuscularity in several organisms. By contrast, Duchenne muscular dystrophy (DMD) is characterized by a loss of muscle fibers and impaired regeneration. In this study, we aim to knockdown myostatin by means of exon skipping, a technique which has been successfully applied to reframe the genetic defect of dystrophin gene in DMD patients. METHODS: We targeted myostatin exon 2 using antisense oligonucleotides (AON) in healthy and DMD-derived myotubes cultures. We assessed the exon skipping level, transcriptional expression of myostatin and its target genes, and combined myostatin and several dystrophin AONs. These AONs were also applied in the mdx mice models via intramuscular injections. RESULTS: Myostatin AON induced exon 2 skipping in cell cultures and to a lower extent in the mdx mice. It was accompanied by decrease in myostatin mRNA and enhanced MYOG and MYF5 expression. Furthermore, combination of myostatin and dystrophin AONs induced simultaneous skipping of both genes. CONCLUSIONS: We conclude that two AONs can be used to target two different genes, MSTN and DMD, in a straightforward manner. Targeting multiple ligands of TGF-beta family will be more promising as adjuvant therapies for DMD.


Assuntos
Distrofina/genética , Éxons/genética , Distrofia Muscular de Duchenne/genética , Miostatina/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos mdx , Desenvolvimento Muscular/efeitos dos fármacos , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular Animal/genética , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Mioblastos/patologia , Miostatina/metabolismo , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
17.
Neurobiol Dis ; 41(2): 353-60, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20940052

RESUMO

Duchenne Muscular Dystrophy (DMD) is an X-linked lethal muscle wasting disease characterized by muscle fiber degeneration and necrosis. The progressive pathology of DMD can be explained by an insufficient regenerative response resulting in fibrosis and adipose tissue formation. BMPs are known to inhibit myogenic differentiation and in a previous study we found an increased expression of a BMP family member BMP4 in DMD myoblasts. The aim of the current study was therefore to investigate whether inhibition of BMP signaling could be beneficial for myoblast differentiation and muscle regeneration processes in a DMD context. All tested BMP inhibitors, Noggin, dorsomorphin and LDN-193189, were able to accelerate and enhance myogenic differentiation. However, dorsomorphin repressed both BMP and TGFß signaling and was found to be toxic to primary myoblast cell cultures. In contrast, Noggin was found to be a potent and selective BMP inhibitor and was therefore tested in vivo in a DMD mouse model. Local adenoviral-mediated overexpression of Noggin in muscle resulted in an increased expression of the myogenic regulatory genes Myog and Myod1 and improved muscle histology. In conclusion, our results suggest that repression of BMP signaling may constitute an attractive adjunctive therapy for DMD patients.


Assuntos
Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Mioblastos/patologia , Fenótipo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/farmacologia , Proteínas de Transporte/uso terapêutico , Diferenciação Celular/genética , Linhagem Celular , Humanos , Masculino , Camundongos , Camundongos Endogâmicos mdx , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo
18.
Pigment Cell Melanoma Res ; 21(3): 379-87, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18444963

RESUMO

Tbx3, a member of the conserved family of T-box developmental transcription factors, is a transcriptional repressor required during cardiogenesis for the formation and specification of the sinoatrial node, the pacemaker of the heart. Both the TBX3 and the highly related TBX2 genes are also associated with several cancers, most likely as a consequence of their powerful anti-senescence properties mediated via suppression p14(Arf) and p21(CIP) expression. In melanoma, the TBX2 gene is frequently amplified and inhibition of Tbx2 function leads to senescence and up-regulation of p21(CIP), a Tbx2 target gene. Tbx3 + 2a is a splice variant containing an extra 20 amino acids encoded by exon 2a inserted into the highly conserved T-box DNA-binding domain. We find here that Tbx3 + 2a is evolutionary conserved and that similar insertions are largely absent from the T-box domains of other T-box factors. Tbx3 + 2a has been reported to lack DNA-binding ability and act as a functional antagonist of Tbx3. By contrast, we now demonstrate that both Tbx3 and Tbx3 + 2a bind the consensus T-element, the p21(CIP1) promoter, and the Nppa cardiac target gene. Both isoforms also function as repressors of p21(CIP1) and Nppa promoter activity and interact with homeobox factor Nkx2-5. When ectopically expressed in the embryonic heart of mice, Tbx3 and Tbx3 + 2a both suppressed chamber formation and repressed expression of cardiac chamber markers Nppa and Cx40. The results suggest that in the assays used, Tbx3 and Tbx3 + 2a are functionally equivalent and that like Tbx2, Tbx3 may also function as an anti-senescence factor in melanoma.


Assuntos
Processamento Alternativo/genética , Sequência Conservada , Éxons , Proteínas com Domínio T/genética , Sequência de Aminoácidos , Animais , Fator Natriurético Atrial , Linhagem Celular , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , DNA Recombinante , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Peptídeo Natriurético Tipo C/genética , Regiões Promotoras Genéticas , Precursores de Proteínas/genética , Alinhamento de Sequência , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/metabolismo
19.
Genes Dev ; 21(9): 1098-112, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17473172

RESUMO

The sinoatrial node initiates the heartbeat and controls the rate and rhythm of contraction, thus serving as the pacemaker of the heart. Despite the crucial role of the sinoatrial node in heart function, the mechanisms that underlie its specification and formation are not known. Tbx3, a transcriptional repressor required for development of vertebrates, is expressed in the developing conduction system. Here we show that Tbx3 expression delineates the sinoatrial node region, which runs a gene expression program that is distinct from that of the bordering atrial cells. We found lineage segregation of Tbx3-negative atrial and Tbx3-positive sinoatrial node precursor cells as soon as cardiac cells turn on the atrial gene expression program. Tbx3 deficiency resulted in expansion of expression of the atrial gene program into the sinoatrial node domain, and partial loss of sinoatrial node-specific gene expression. Ectopic expression of Tbx3 in mice revealed that Tbx3 represses the atrial phenotype and imposes the pacemaker phenotype on the atria. The mice displayed arrhythmias and developed functional ectopic pacemakers. These data identify a Tbx3-dependent pathway for the specification and formation of the sinoatrial node, and show that Tbx3 regulates the pacemaker gene expression program and phenotype.


Assuntos
Função Atrial/genética , Nó Sinoatrial/embriologia , Nó Sinoatrial/fisiologia , Proteínas com Domínio T/genética , Animais , Sequência de Bases , Diferenciação Celular , Primers do DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Átrios do Coração/citologia , Átrios do Coração/embriologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/fisiologia , Nó Sinoatrial/citologia , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/fisiologia
20.
Circ Res ; 100(3): 354-62, 2007 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-17234970

RESUMO

The sinoatrial node, which resides at the junction of the right atrium and the superior caval vein, contains specialized myocardial cells that initiate the heart beat. Despite this fundamental role in heart function, the embryonic origin and mechanisms of localized formation of the sinoatrial node have not been defined. Here we show that subsequent to the formation of the Nkx2-5-positive heart tube, cells bordering the inflow tract of the heart tube give rise to the Nkx2-5-negative myocardial cells of the sinoatrial node and the sinus horns. Using genetic models, we show that as the myocardium of the heart tube matures, Nkx2-5 suppresses pacemaker channel gene Hcn4 and T-box transcription factor gene Tbx3, thereby enforcing a progressive confinement of their expression to the forming Nkx2-5-negative sinoatrial node and sinus horns. Thus, Nkx2-5 is essential for establishing a gene expression border between the atrium and sinoatrial node. Tbx3 was found to suppress chamber differentiation, providing an additional mechanism by which the Tbx3-positive sinoatrial node is shielded from differentiating into atrial myocardium. Pitx2c-deficient fetuses form sinoatrial nodes with indistinguishable molecular signatures at both the right and left sinuatrial junction, indicating that Pitx2c functions within the left/right pathway to suppress a default program for sinuatrial node formation on the left. Our molecular pathway provides a mechanism for how pacemaker activity becomes progressively relegated to the most recently added components of the venous pole of the heart and, ultimately, to the junction of the right atrium and superior caval vein.


Assuntos
Padronização Corporal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Átrios do Coração/embriologia , Ventrículos do Coração/embriologia , Proteínas de Homeodomínio/fisiologia , Canais Iônicos/biossíntese , Nó Sinoatrial/embriologia , Proteínas com Domínio T/fisiologia , Fatores de Transcrição/fisiologia , Animais , Fator Natriurético Atrial , Biomarcadores , Padronização Corporal/genética , Miosinas Cardíacas/biossíntese , Miosinas Cardíacas/genética , Conexinas/biossíntese , Conexinas/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Reporter , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Imageamento Tridimensional , Hibridização In Situ , Canais Iônicos/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miocárdio/metabolismo , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/genética , Peptídeo Natriurético Tipo C/biossíntese , Peptídeo Natriurético Tipo C/genética , Precursores de Proteínas/biossíntese , Precursores de Proteínas/genética , Proteínas Recombinantes de Fusão/fisiologia , Nó Sinoatrial/citologia , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Troponina I/biossíntese , Troponina I/genética , Proteína alfa-5 de Junções Comunicantes , Proteína Homeobox PITX2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...