Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Free Radic Biol Med ; 129: 364-371, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30312761

RESUMO

Generation of superoxide by xanthine oxidase can be stimulated under ischemic and aberrant calcium homeostasis. Because patients and mice with Duchenne muscular dystrophy (DMD) suffer from ischemia and excessive calcium influx, we tested the hypothesis that xanthine oxidase activity is elevated and contributes to disease pathology. Xanthine oxidase activity was measured by urinary isoxanthopterin in DMD patients at rest and in response to exercise. Urinary isoxanthopterin/creatinine was elevated compared to age-matched controls and Becker muscular dystrophy (BMD) patients. Concentrations were also increased after a six minute walk test in ambulatory patients. We also measured urinary isoxanthopterin in wildtype mice and a number of dystrophic mouse models; the DMD mouse model (mdx), mdx mice overexpressing a variety of transgenic miniaturized and chimeric skeletal muscle-specific dystrophins and utrophin and the ß-sarcoglycan deficient (Scgb-/-) mouse which represents type 2E human limb-girdle muscular dystrophy. Mdx and Scgb-/-mice had greater urinary isoxanthopterin/creatinine than wildtype mice while mdx mice expressing dystrophin or utrophin linking the extracellular matrix to the actin cytoskeleton were not different than wildtype. We also measured higher levels of urinary ortho-tyrosine in humans and mice deficient for dystrophin to confirm elevated oxidative stress. Surprisingly, mdx had lower xanthine oxidase protein levels and higher mRNA in gastrocnemius muscle compared to wildtype mice, however, the enzymatic activity of skeletal muscle xanthine oxidase was elevated above wildtype and a transgenic rescued mdx mouse (DysΔMTB-mdx). Downhill treadmill running also caused significant increases in mdx urinary isoxanthopterin that was prevented with the xanthine oxidase inhibitor allopurinol. Similarly, in vitro eccentric contraction-induced force drop of mdx muscle was attenuated by the allopurinol metabolite, oxypurinol. Together, our data suggests hyper-activity of xanthine oxidase in DMD, identifies xanthine oxidase activity as a contributing factor in eccentric contraction-induced force drop of dystrophin-deficient skeletal muscle and highlights the potential of isoxanthopterin as a noninvasive biomarker in DMD.


Assuntos
Distrofina/deficiência , Distrofia Muscular Animal/enzimologia , Distrofia Muscular de Duchenne/enzimologia , Xantina Oxidase/urina , Xantopterina/urina , Adolescente , Alopurinol/farmacologia , Animais , Biomarcadores/urina , Estudos de Casos e Controles , Creatinina/urina , Distrofina/genética , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos mdx , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/enzimologia , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/tratamento farmacológico , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatologia , Oxipurinol/farmacologia , Sarcoglicanas/deficiência , Sarcoglicanas/genética , Tirosina/urina , Utrofina/deficiência , Utrofina/genética , Xantina Oxidase/genética , Adulto Jovem
2.
J Biol Chem ; 291(19): 9920-8, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-26966179

RESUMO

Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by mutations in the gene encoding dystrophin. Loss of dystrophin protein compromises the stability of the sarcolemma membrane surrounding each muscle cell fiber, leading to membrane ruptures and leakiness that induces myofiber necrosis, a subsequent inflammatory response, and progressive tissue fibrosis with loss of functional capacity. Cathepsin S (Ctss) is a cysteine protease that is actively secreted in areas of tissue injury and ongoing inflammation, where it participates in extracellular matrix remodeling and healing. Here we show significant induction of Ctss expression and proteolytic activity following acute muscle injury or in muscle from mdx mice, a model of DMD. To examine the functional ramifications associated with greater Ctss expression, the Ctss gene was deleted in the mdx genetic background, resulting in protection from muscular dystrophy pathogenesis that included reduced myofiber turnover and histopathology, reduced fibrosis, and improved running capacity. Mechanistically, deletion of the Ctss gene in the mdx background significantly increased myofiber sarcolemmal membrane stability with greater expression and membrane localization of utrophin, integrins, and ß-dystroglycan, which anchor the membrane to the basal lamina and underlying cytoskeletal proteins. Consistent with these results, skeletal muscle-specific transgenic mice overexpressing Ctss showed increased myofiber necrosis, muscle histopathology, and a functional deficit reminiscent of muscular dystrophy. Hence, Ctss induction during muscular dystrophy is a pathologic event that partially underlies disease pathogenesis, and its inhibition might serve as a new therapeutic strategy in DMD.


Assuntos
Catepsinas/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Fibras Musculares Esqueléticas/enzimologia , Distrofia Muscular Animal/enzimologia , Distrofia Muscular de Duchenne/enzimologia , Animais , Citoesqueleto/enzimologia , Citoesqueleto/genética , Citoesqueleto/patologia , Camundongos , Camundongos Endogâmicos mdx , Camundongos Knockout , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Necrose , Proteólise , Sarcolema/enzimologia , Sarcolema/genética , Sarcolema/patologia
3.
Am J Pathol ; 184(6): 1819-30, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24726497

RESUMO

Abnormalities in phosphoinositide metabolism are an emerging theme in human neurodegenerative disease. Myotubular myopathy is a prototypical disorder of phosphoinositide dysregulation that is characterized by profound muscle pathology and weakness and that is caused by mutations in MTM1, which encodes a phosphatase that targets 3-position phosphoinositides, including phosphatidylinositol 3-phosphate. Although the association between MTM1 and muscle disease has become increasingly clarified, the normal role(s) of phosphatidylinositol 3-phosphate metabolism in muscle development and homeostasis remain poorly understood. To begin to address the function of phosphatidylinositol 3-phosphate in skeletal muscle, we focused on the primary kinase responsible for its production, and created a muscle-specific conditional knockout of the class III phosphatidylinositol 3-kinase, Pik3c3. Muscle-specific deletion of Pik3c3 did not disturb embryogenesis or early postnatal development, but resulted in progressive disease characterized by reduced activity and death by 2 months of age. Histopathological analysis demonstrated changes consistent with a murine muscular dystrophy. Examination for cellular mechanism(s) responsible for the dystrophic phenotype revealed significant alterations in the autophagolysosomal pathway with mislocation of known dystrophy proteins to the lysosomal compartment. In all, we present the first analysis of Pik3c3 in skeletal muscle, and report a novel association between deletion of Pik3c3 and muscular dystrophy.


Assuntos
Autofagia , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/enzimologia , Fosfatos de Fosfatidilinositol/metabolismo , Animais , Classe III de Fosfatidilinositol 3-Quinases/genética , Lisossomos/metabolismo , Lisossomos/patologia , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Músculo Esquelético/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Fosfatos de Fosfatidilinositol/genética
4.
Hum Gene Ther ; 23(1): 98-103, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21933029

RESUMO

Six- to 8-kb mini-dystrophin genes are promising candidates for Duchenne muscular dystrophy (DMD) gene therapy. Several dual adeno-associated virus (AAV) mini-dystrophin vectors have been tested in dystrophin-deficient mice. Despite the encouraging preclinical results, none of the existing dual AAV vectors can restore sarcolemmal neuronal nitric oxide synthase (nNOS) expression. Localization of nNOS to the sarcolemma may greatly improve the therapeutic outcome in DMD (Lai, Y., Thomas, G.D., Yue, Y., et al. [2009]. J. Clin. Invest. 119, 624-635). In this study, we developed a series of dual AAV expression vectors to express a synthetic minigene that carries the nNOS localization domain. To help validate dual vector reconstitution, we also included a FLAG tag and a GFP reporter at different ends of the minigene. These dual AAV vectors were packaged in Y445F tyrosine mutant AAV-6 and tested in dystrophin-null mdx4cv mice by direct muscle injection. All dual vectors expressed GFP/FLAG-tagged mini-dystrophin and restored sarcolemmal nNOS. However, the reconstitution efficiency was significantly different among different sets. The dual vector set YZ27/YZ22 yielded the highest transduction efficiency (∼90%). Further development of this set dual vector may lead to more effective DMD gene therapy.


Assuntos
Dependovirus/metabolismo , Distrofina/metabolismo , Vetores Genéticos/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Sarcolema/enzimologia , Animais , Western Blotting , Dependovirus/genética , Distrofina/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Injeções Intramusculares , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/citologia , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/enzimologia , Distrofia Muscular Animal/patologia , Distrofia Muscular Animal/terapia , Óxido Nítrico Sintase Tipo I/genética , Plasmídeos/genética , Plasmídeos/metabolismo , Sarcolema/patologia
5.
J Neurol Sci ; 303(1-2): 53-60, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21306738

RESUMO

To examine potential mechanisms for the reduced resting membrane potentials (RPs) of mature dystrophic (mdx) muscle fibers, the Na(+)-K(+) pump inhibitor ouabain was added to freshly isolated nondystrophic and mdx fibers. Ouabain produced a 71% smaller depolarization in mdx fibers than in nondystrophic fibers, increased the [Na(+)](i) in nondystrophic fibers by 40%, but had no significant effect on the [Na(+)](i) of mdx fibers, which was approximately double that observed in untreated nondystrophic fibers. Western blots indicated no difference in total and phosphorylated Na(+)-K(+) ATPase catalytic α1 subunit between nondystrophic and mdx muscle. Examination of the effects of the NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) indicated that direct application of the drug slowly hyperpolarized mdx fibers (7 mV in 90 min) but had no effect on nondystrophic fibers. Pretreatment with ouabain abolished this hyperpolarization, and pretreatment with PDTC restored ouabain-induced depolarization and reduced [Na(+)](i). Administration of an NF-κB inhibitor that utilizes a different mechanism for reducing nuclear NF-κB activation, ursodeoxycholic acid (UDCA), also hyperpolarized mdx fibers. These results suggest that in situ Na(+)-K(+) pump activity is depressed in mature dystrophic fibers by NF-κB dependent modulators, and that this reduced pump activity contributes to the weakness characteristic of dystrophic muscle.


Assuntos
Inibidores Enzimáticos/farmacologia , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular Animal/patologia , NF-kappa B/fisiologia , Ouabaína/farmacologia , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Animais , Western Blotting , DNA/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Microeletrodos , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Animal/enzimologia , Distrofia Muscular Animal/metabolismo , Pirrolidinas/farmacologia , Sódio/metabolismo , Tiocarbamatos/farmacologia , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/biossíntese , Fator de Transcrição RelA/genética , Ácido Ursodesoxicólico/farmacologia
6.
PLoS One ; 5(5): e10763, 2010 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-20505827

RESUMO

BACKGROUND: Duchenne muscular dystrophy (DMD) is the most common, lethal disease of childhood. One of 3500 new-born males suffers from this universally-lethal disease. Other than the use of corticosteroids, little is available to affect the relentless progress of the disease, leading many families to use dietary supplements in hopes of reducing the progression or severity of muscle wasting. Arginine is commonly used as a dietary supplement and its use has been reported to have beneficial effects following short-term administration to mdx mice, a genetic model of DMD. However, the long-term effects of arginine supplementation are unknown. This lack of knowledge about the long-term effects of increased arginine metabolism is important because elevated arginine metabolism can increase tissue fibrosis, and increased fibrosis of skeletal muscles and the heart is an important and potentially life-threatening feature of DMD. METHODOLOGY: We use both genetic and nutritional manipulations to test whether changes in arginase metabolism promote fibrosis and increase pathology in mdx mice. Our findings show that fibrotic lesions in mdx muscle are enriched with arginase-2-expressing macrophages and that muscle macrophages stimulated with cytokines that activate the M2 phenotype show elevated arginase activity and expression. We generated a line of arginase-2-null mutant mdx mice and found that the mutation reduced fibrosis in muscles of 18-month-old mdx mice, and reduced kyphosis that is attributable to muscle fibrosis. We also observed that dietary supplementation with arginine for 17-months increased mdx muscle fibrosis. In contrast, arginine-2 mutation did not reduce cardiac fibrosis or affect cardiac function assessed by echocardiography, although 17-months of dietary supplementation with arginine increased cardiac fibrosis. Long-term arginine treatments did not decrease matrix metalloproteinase-2 or -9 or increase the expression of utrophin, which have been reported as beneficial effects of short-term treatments. CONCLUSIONS/SIGNIFICANCE: Our findings demonstrate that arginine metabolism by arginase promotes fibrosis of muscle in muscular dystrophy and contributes to kyphosis. Our findings also show that long-term, dietary supplementation with arginine exacerbates fibrosis of dystrophic heart and muscles. Thus, commonly-practiced dietary supplementation with arginine by DMD patients has potential risk for increasing pathology when performed for long periods, despite reports of benefits acquired with short-term supplementation.


Assuntos
Arginina/metabolismo , Macrófagos/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Animal/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Animais , Arginase/metabolismo , Arginina/administração & dosagem , Arginina/farmacologia , Cardiomiopatia Dilatada/enzimologia , Cardiomiopatia Dilatada/patologia , Citocinas/metabolismo , Distrofina/deficiência , Distrofina/metabolismo , Fibrose , Deleção de Genes , Inflamação/complicações , Inflamação/enzimologia , Inflamação/patologia , Cifose/complicações , Cifose/enzimologia , Cifose/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/complicações , Distrofia Muscular Animal/enzimologia , Óxido Nítrico Sintase Tipo I/metabolismo , Transporte Proteico/efeitos dos fármacos , Células Th2/efeitos dos fármacos
7.
J Cell Sci ; 123(Pt 6): 960-71, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20179101

RESUMO

Here, we characterise new strains of normal and dystrophic (mdx) mice that overexpress Class 2 IGF-1 Ea in skeletal myofibres. We show that transgenic mice have increased muscle levels of IGF-1 (approximately 13-26 fold) and show striking muscle hypertrophy (approximately 24-56% increase in mass). Adult normal muscles were resistant to elevated IGF-1; they reached adult steady state and maintained the same mass from 3 to 12 months. By contrast, dystrophic muscles from mdx/IGF-1(C2:Ea) mice continued to increase in mass during adulthood. IGF-1 signalling was evident only in muscles that were growing as a result of normal postnatal development (23-day-old mice) or regenerating in response to endogenous necrosis (adult mdx mice). Increased phosphorylation of Akt at Ser473 was not evident in fasted normal adult transgenic muscles, but was 1.9-fold higher in fasted normal young transgenic muscles compared with age-matched wild-type controls and fourfold higher in fasted adult mdx/IGF-1(C2:Ea) compared with mdx muscles. Muscles of adult mdx/IGF-1(C2:Ea) mice showed higher p70(S6K)(Thr421/Ser424) phosphorylation and both young transgenic and adult mdx/IGF-1(C2:Ea) mice had higher phosphorylation of rpS6(Ser235/236). The level of mRNA encoding myogenin was increased in normal young (but not adult) transgenic muscles, indicating enhanced myogenic differentiation. These data demonstrate that elevated IGF-1 has a hypertrophic effect on skeletal muscle only in growth situations.


Assuntos
Fator de Crescimento Insulin-Like I/metabolismo , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/patologia , Envelhecimento/patologia , Animais , Peso Corporal , Regulação da Expressão Gênica , Hipertrofia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Endogâmicos mdx , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/sangue , Distrofia Muscular Animal/enzimologia , Distrofia Muscular Animal/patologia , Miogenina/genética , Miogenina/metabolismo , Tamanho do Órgão , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR
8.
J Cell Biol ; 187(6): 859-74, 2009 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-20008564

RESUMO

Mammalian target of rapamycin (mTOR) is a key regulator of cell growth that associates with raptor and rictor to form the mTOR complex 1 (mTORC1) and mTORC2, respectively. Raptor is required for oxidative muscle integrity, whereas rictor is dispensable. In this study, we show that muscle-specific inactivation of mTOR leads to severe myopathy, resulting in premature death. mTOR-deficient muscles display metabolic changes similar to those observed in muscles lacking raptor, including impaired oxidative metabolism, altered mitochondrial regulation, and glycogen accumulation associated with protein kinase B/Akt hyperactivation. In addition, mTOR-deficient muscles exhibit increased basal glucose uptake, whereas whole body glucose homeostasis is essentially maintained. Importantly, loss of mTOR exacerbates the myopathic features in both slow oxidative and fast glycolytic muscles. Moreover, mTOR but not raptor and rictor deficiency leads to reduced muscle dystrophin content. We provide evidence that mTOR controls dystrophin transcription in a cell-autonomous, rapamycin-resistant, and kinase-independent manner. Collectively, our results demonstrate that mTOR acts mainly via mTORC1, whereas regulation of dystrophin is raptor and rictor independent.


Assuntos
Proteínas de Transporte/metabolismo , Distrofina/metabolismo , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Fatores Etários , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Células Cultivadas , Distrofina/genética , Eletroporação , Metabolismo Energético , Ativação Enzimática , Feminino , Glucose/metabolismo , Glicogênio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Musculares/enzimologia , Contração Muscular , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Mutação , Oxirredução , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina , Ratos , Proteína Regulatória Associada a mTOR , Índice de Gravidade de Doença , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Transdução Genética , Utrofina/metabolismo
9.
Am J Pathol ; 174(5): 1735-44, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19359520

RESUMO

Duchenne muscular dystrophy is a fatal muscle wasting disease that is characterized by a deficiency in the protein dystrophin. Previously, we reported that the expression of hematopoietic prostaglandin D synthase (HPGDS) appeared in necrotic muscle fibers from patients with either Duchenne muscular dystrophy or polymyositis. HPGDS is responsible for the production of the inflammatory mediator, prostaglandin D(2). In this paper, we validated the hypothesis that HPGDS has a role in the etiology of muscular necrosis. We investigated the expression of HPGDS/ prostaglandin D(2) signaling using two different mouse models of muscle necrosis, that is, bupivacaine-induced muscle necrosis and the mdx mouse, which has a genetic muscular dystrophy. We treated each mouse model with the HPGDS-specific inhibitor, HQL-79, and measured both necrotic muscle volume and selected cytokine mRNA levels. We confirmed that HPGDS expression was induced in necrotic muscle fibers in both bupivacaine-injected muscle and mdx mice. After administration of HQL-79, necrotic muscle volume was significantly decreased in both mouse models. Additionally, mRNA levels of both CD11b and transforming growth factor beta1 were significantly lower in HQL-79-treated mdx mice than in vehicle-treated animals. We also demonstrated that HQL-79 suppressed prostaglandin D(2) production and improved muscle strength in the mdx mouse. Our results show that HPGDS augments inflammation, which is followed by muscle injury. Furthermore, the inhibition of HPGDS ameliorates muscle necrosis even in cases of genetic muscular dystrophy.


Assuntos
Modelos Animais de Doenças , Oxirredutases Intramoleculares/antagonistas & inibidores , Lipocalinas/antagonistas & inibidores , Distrofia Muscular Animal/prevenção & controle , Anestésicos Locais/toxicidade , Animais , Western Blotting , Bupivacaína/toxicidade , Citocinas/genética , Humanos , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Lipocalinas/genética , Lipocalinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Camundongos Transgênicos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Distrofia Muscular Animal/induzido quimicamente , Distrofia Muscular Animal/enzimologia , Necrose , Piperidinas/farmacologia , Prostaglandina D2/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
J Mol Neurosci ; 30(1-2): 49-50, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17192623

RESUMO

Lama2dy mice constitute an animal model for congenital muscular dystrophy (CMD) by merosin (laminin alpha2-chain) deficiency. This pathology affects the properties of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) of mouse skeletal muscle and nerves (Moral-Naranjo et al., 1999, 2002). AChE and BChE are involved through catalytic and noncatalytic actions in multiple processes, such as hydrolysis of acetylcholine (ACh), morphogenesis, hematopoiesis, and tumorigenesis (Soreq and Seidman, 2001). AChE and BChE molecules can be globular (G1, G2, and G4) or asymmetric forms (A4, A8, and A12) (Massoulié, 2002), and G molecules can show amphiphilic (detergent-interacting, GA) or hydrophilic (GH) behavior. AChE catalytic subunits are encoded by three mRNAs (T, H, or R) generated by alternative splicing. The presence of AChE in lymphoid tissues (Rossi et al., 1991; Nieto-Cerón et al., 2004), the role of immune responses in muscular dystrophy (Spencer and Tidball, 2001), the abnormalities of Lama2dy thymus (Magner et al., 2000), and the role of ACh in thymocyte function (Kawashima and Fujii, 2000) prompted us to investigate thymus AChE and the possible effect of merosin deficiency on it.


Assuntos
Acetilcolinesterase/metabolismo , Distrofia Muscular Animal/enzimologia , Timo/metabolismo , Animais , Isoenzimas/metabolismo , Laminina/deficiência , Linfócitos/enzimologia , Camundongos , Camundongos Endogâmicos , Valores de Referência
11.
Bioorg Med Chem Lett ; 15(23): 5176-81, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16185867

RESUMO

Dipeptide-derived alpha-keto-amide compounds with potent calpain inhibitory activity have been identified. These reversible covalent inhibitors have IC(50) values down to 25nM and exhibit greatly improved activity in muscle cells compared to the reference compound MDL28170. Several novel calpain inhibitors have shown positive effects on histological parameters in an animal model of Duchenne muscular dystrophy demonstrating their potential as a treatment option for this fatal disease.


Assuntos
Amidas/uso terapêutico , Calpaína/antagonistas & inibidores , Inibidores de Cisteína Proteinase/química , Inibidores de Cisteína Proteinase/uso terapêutico , Dipeptídeos/química , Distrofia Muscular Animal/tratamento farmacológico , Amidas/química , Animais , Diafragma/efeitos dos fármacos , Diafragma/patologia , Camundongos , Camundongos Endogâmicos mdx , Estrutura Molecular , Distrofia Muscular Animal/enzimologia
12.
J Neurosci Res ; 66(4): 656-65, 2001 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11746385

RESUMO

Ecto-5'-nucleotidase (eNT) activity and protein in normal (NM) and merosin-deficient dystrophic (DM) Lama2(dy) mice muscle were studied. eNT activity in DM was three- to four-fold that in NM. eNT in NM and DM displayed the same kinetic properties. Slot and Western blotting revealed that the immunoreactive protein was two to three times more abundant in control muscle, when NM and DM samples with the same eNT activity were compared, indicating that mouse muscle contains catalytically inactive eNT components. eNT activity and protein peaks coincided in sedimentation analyses, revealing that inactive eNT occurs as dimers. Most eNT activity and protein of NM bound to Lens culinaris (LCA) or Ricinus communis (RCA) agglutinins, but half of the activity and one-third of the protein bound to wheat germ agglutinin (WGA). Although WGA interaction did not permit full separation of inactive eNT, the results suggest that similar proportions of active species with and without WGA reactivity occur in mouse muscle, whereas a great fraction of the inactive eNT variants lack WGA reactivity. Because the level of eNT protein was little modified in DM, the higher eNT activity in dystrophic than in control muscle may result from misregulation in the synthesis of active and inactive eNT species or from conversion of inactive into active components.


Assuntos
5'-Nucleotidase/metabolismo , Laminina/deficiência , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/enzimologia , Monofosfato de Adenosina/metabolismo , Animais , Interações Hidrofóbicas e Hidrofílicas , Cinética , Laminina/genética , Lectinas/química , Camundongos , Camundongos Mutantes , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Frações Subcelulares , Tensoativos/metabolismo
13.
Curr Biol ; 11(16): 1278-82, 2001 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-11525743

RESUMO

Duchenne Muscular Dystrophy (DMD) originates from deleterious mutations in the dystrophin gene, with a complete loss of the protein product. Subsequently, the disease is manifested in severe striated muscle wasting and death in early adulthood. Dystrophin provides a structural base for the assembly of an integral membrane protein complex. As such, dystrophin deficiency leads to an altered mechanical integrity of the myofiber and a predisposition to contraction-induced damage. However, the development of myofiber degeneration prior to an observed mechanical defect has been documented in various dystrophic models. Although activation of a detrimental signal transduction pathway has been suggested as a probable cause, a specific cellular cascade has yet to be defined. Here, it is shown that murine models of DMD displayed a muscle-specific activation of JNK1. Independent activation of JNK1 resulted in defects in myotube viability and integrity in vitro, similar to a dystrophic phenotype. In addition, direct muscle injection of an adenoviral construct containing the JNK1 inhibitory protein, JIP1, dramatically attenuated the progression of dystrophic myofiber destruction. Taken together, these results suggest that a JNK1-mediated signal cascade is a conserved feature of dystrophic muscle and contributes to the progression of the disease pathogenesis.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/enzimologia , Distrofia Muscular de Duchenne/patologia , Adenoviridae/genética , Animais , Células Cultivadas , Ativação Enzimática , Proteínas de Fluorescência Verde , Humanos , Indicadores e Reagentes/metabolismo , Proteínas Luminescentes/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/genética , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/enzimologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/genética , Miocárdio/enzimologia , Miocárdio/metabolismo , Miocárdio/patologia , Fosforilação , Transfecção
14.
Dev Biol ; 205(1): 158-70, 1999 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-9882504

RESUMO

Matrix metalloproteinases (MMPs) cooperatively degrade all components of the extracellular matrix (ECM). Remodeling of ECM during skeletal muscle degeneration and regeneration suggests a tight regulation of matrix-degrading activity during muscle regeneration. In this study, we investigated the expression of MMP-2 and MMP-9, in normal muscles and their regulation during regeneration process. We further investigated their secretion by C2C12 myogenic cell line. Two models of muscle degeneration-regeneration were used: (1) normal muscles in which necrosis was experimentally induced by cardiotoxin injection; (2) mdx muscles which exhibit recurrent signs of focal myofiber necrosis followed by successful regeneration. MMPs were studied by zymography; their free activity was quantified using 3H-labeled gelatin substrate and mRNA expression was followed by Northern hybridization. Muscle degeneration-regeneration was analyzed by conventional morphological methods and in situ hybridization was performed on muscle sections to identify the cells expressing these MMPs. Results show that MMP-2, but not MMP-9 expression, is constitutive in normal muscles. Upon injury, the active form of MMP-2 is transiently increased, whereas MMP-9 is induced within 24 h and remains present for several days. Quantitative assays of free gelatinolytic activity show a progressive and steady increase that culminates at 7 days postinjury and slowly returns to normal levels. In adult mdx mice, both pro and active forms of MMP-2 and MMP-9 are expressed. Northern blot results support these findings. Zymography of C2C12-conditioned medium shows that myogenic cells produce MMP-2. By in situ hybridization we localized MMP-9 mRNA in inflammatory cells and putative activated satellite cells in injured muscles. Our data allow the correlation of the differential expression of pro and/or active forms of MMP-2 and MMP-9 with different stages of the degeneration-regeneration process: MMP-9 expression is related to the inflammatory response and probably to the activation of satellite cells, whereas MMP-2 activation is concomitant with the regeneration of new myofibers.


Assuntos
Colagenases/genética , Gelatinases/genética , Regulação Enzimológica da Expressão Gênica , Metaloendopeptidases/genética , Músculo Esquelético/fisiologia , Distrofia Muscular Animal/fisiopatologia , Regeneração , Animais , Linhagem Celular , Proteínas Cardiotóxicas de Elapídeos , Metaloproteinase 2 da Matriz , Metaloproteinase 9 da Matriz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculo Esquelético/lesões , Músculo Esquelético/patologia , Distrofia Muscular Animal/enzimologia , Necrose
15.
Gene Ther ; 5(1): 59-64, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9536265

RESUMO

In normal skeletal muscle fibers, dystrophin accumulates at the cytoplasmic face of the sarcolemma where it associates with dystrophin-associated proteins (DAPs). Several studies have recently shown that the neuronal isoform of nitric oxide synthase (nNOS) is also located at the sarcolemma, and that this membrane localization is mediated through interactions of nNOS with one of the DAPs, namely alpha 1-syntrophin. Since the lack of dystrophin in muscle fibers from Duchenne muscular dystrophy patients and mdx mice is accompanied by an absence of sarcolemmal nNOS, we examined in the present study, whether dystrophin gene replacement would lead to the restoration of nNOS at its appropriate subcellular location. To this end, tibialis anterior muscles from mdx4cv mice were directly injected with plasmid DNA encoding either full-length (pRSV-dys) or mini-(pRSV-dyB; lacking exons 17-48) dystrophin. For these experiments, we chose to study 10-week-old mdx4cv mice since at this developmental stage, muscles from these mice have already undergone several cycles of degeneration-regeneration. Immunofluorescence experiments performed on serial cross-sections revealed that approximately 50% of the dystrophin-positive fibers also exhibited significant levels of nNOS at their sarcolemma 2 weeks following gene transfer with pRSV-dys. Similar results were obtained with pRSV-dyB indicating that exons 17-48 of the dystrophin gene are not essential for the correct localization of nNOS in skeletal muscle fibers. Taken together with the recent demonstration that dystrophin gene transfer leads to significant physiological benefits our results suggest that dystrophin gene therapy using full-length or truncated dystrophin, also induces a rapid recovery of biochemical functions.


Assuntos
Distrofina/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Distrofia Muscular Animal/terapia , Óxido Nítrico Sintase/metabolismo , Sarcolema/enzimologia , Animais , Vírus do Sarcoma Aviário , Imunofluorescência , Expressão Gênica , Vetores Genéticos , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/enzimologia , Óxido Nítrico Sintase/análise , Plasmídeos
16.
J Neurochem ; 69(5): 1964-74, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9349541

RESUMO

The distribution and glycosylation of acetylcholinesterase (AChE) forms in vesicles derived from sarcoplasmic reticulum of normal muscle (NMV) were investigated and compared with those from dystrophic muscle vesicles (DMV). AChE activity was similar in NMV and DMV. Most of the AChE in NMV and half in DMV were released with Triton X-100. Asymmetric (A12) and globular hydrophilic and amphiphilic (G4H, G4A, G2A, and G1A) AChE species occurred in NMV and DMV, the lighter forms being predominant. The percentage of G4H and G4A decreased in DMV. A fraction of the AChE that could not be extracted with detergent was detached with collagenase. Most of the detergent-released A12 AChE from NMV and nearly half in DMV failed to bind to Ricinus communis agglutinin (RCA-I). Conversely, the collagenase-detached isoforms bound to RCA, revealing that asymmetric AChE associated with internal membranes or basal lamina differed in glycosylation. Moreover, nearly half of G4A AChE in DMV and a few in NMV bound to RCA. Most of the RCA-unreactive G4A forms in NMV come from sarcolemma. The results indicate that dystrophy induces minor changes in the distribution and glycosylation of AChE forms in internal membranes of muscle.


Assuntos
Acetilcolinesterase/metabolismo , Isoenzimas/metabolismo , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/enzimologia , Acetilcolinesterase/isolamento & purificação , Adenosina Trifosfatases/metabolismo , Animais , Butirilcolinesterase/metabolismo , Glicosilação , Membranas Intracelulares/enzimologia , Isoenzimas/isolamento & purificação , Camundongos , Camundongos Mutantes Neurológicos , Microssomos/enzimologia , Modelos Biológicos , Distrofia Muscular Animal/genética , Valores de Referência
17.
Biochim Biophys Acta ; 1290(1): 4-8, 1996 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-8645705

RESUMO

In Duchenne muscular dystrophy, muscle cells, which lack the protein dystrophin, have been reported to have elevated resting intracellular calcium levels. It has also been noted that, compared to normal muscle, intracellular [Ca2+] in dystrophic muscle returns more slowly to its resting level following contractile stimulation. Consistent with this, it has been suggested that dystrophin is directly involved in the regulation of Ca2+ influx. A secondary alteration in the sarcoplasmic reticulum Ca2+ pump, however, could also contribute to, or be responsible for, the abnormal Ca2+ handling seen. To determine whether the Ca2+ pump is functionally altered in dystrophic muscle, we examined Ca2+ uptake by vesicles derived from skeletal muscle sarcoplasmic reticulum of normal and dystrophic (mdx) mice. The Hill coefficient and the Ca2+ sensitivity of the Ca2+- ATPase were the same in both cases. The maximum velocity of Ca2+ uptake, however, normalized to the ATPase content of the vesicles, was less for mdx muscle.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/enzimologia , Retículo Sarcoplasmático/enzimologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL
18.
J Neurol Sci ; 133(1-2): 11-5, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8583212

RESUMO

Recent evidence indicates that in dystrophin-deficient muscle, intracellular sodium content (Na(i)) may be elevated and sodium regulation may be altered or impaired. If there is an elevation in Na(i), this could be due to decreased active pumping of sodium from the cell or increased passive influx of sodium. The present study has therefore determined the content of plasma membrane-bound Na+/K(+)-ATPase in the skeletal muscle of mdx mice; a genetically homologous model of Duchenne muscular dystrophy. Measurements were made on muscles from 5-6-month-old mdx mice and age-matched controls of the C57B1/10ScSn strain (n = 9 pairs), using the vanadate-facilitated ouabain-binding technique. The Na+/K(+)-ATPase concentration per unit weight increased by 2.3-fold in the longissimus dorsi and 1.4-fold in the gastrocnemius of mdx mice compared with controls. The increase in Na+/K(+)-ATPase content is of similar magnitude to the previously reported increase in ouabain-sensitive Na+/K(+)-ATPase activity in mdx muscle, suggesting that this elevated enzyme activity occurs largely through an increase in its concentration. This compensatory increase in the main regulator of internal sodium is likely to occur in an attempt to maintain homeostasis. Nevertheless, the elevated pump concentration is unable to compensate entirely for the increased Na(i). These results are consistent with a previously proposed hypothesis that sodium regulation is abnormal in dystrophin deficient muscles, and also that cell death in these muscles may be due to abnormal regulation of cell volume.


Assuntos
Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/enzimologia , Ouabaína/farmacologia , ATPase Trocadora de Sódio-Potássio/efeitos dos fármacos , Sódio/metabolismo , Animais , Distrofina/deficiência , Técnicas In Vitro , Camundongos , Camundongos Mutantes Neurológicos , Músculo Esquelético/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo
19.
Am J Physiol ; 265(3 Pt 1): C834-41, 1993 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8214039

RESUMO

The X chromosome-linked muscular dystrophic (mdx) mouse lacks the subsarcolemmal protein dystrophin and thus represents a genetic homologue of human Duchenne muscular dystrophy. The present study examined alterations in diaphragm contractile properties and myosin heavy chain (MHC) expression in young (3-4 mo) and old (22-24 mo) control and mdx mice. In young mdx mice, maximum isometric tension (Po) was reduced to 50% of control values. An increase in fibers coexpressing types I (slow) and IIa MHC as well as regenerating fibers expressing embryonic MHC occurred, whereas IIx/b fibers were decreased. In the old mdx group, Po underwent a further reduction to 25% of control, and there was a slowing of twitch kinetics along with markedly increased diaphragm endurance. These changes were associated with an approximate sevenfold increase in type I MHC fibers and virtual elimination of the IIx/b fiber population; there was no detectable embryonic MHC expression. We conclude that the mdx diaphragm responds to progressive muscle degeneration with transition to a slower phenotype associated with reduced power output and augmented muscle endurance. In the setting of progressive muscle fiber destruction, these changes may help preserve contractile function and promote greater survival of remaining muscle fibers by decreasing cellular energy requirements.


Assuntos
Adaptação Fisiológica , Diafragma/fisiopatologia , Contração Muscular , Distrofia Muscular Animal/enzimologia , Distrofia Muscular Animal/fisiopatologia , Miosinas/metabolismo , Animais , Diafragma/enzimologia , Diafragma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Mutantes , Distrofia Muscular Animal/patologia , Miosinas/química
20.
Int J Biochem ; 24(4): 579-84, 1992 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1355451

RESUMO

1. Experiments were carried out to examine the biochemical changes, such as contractile protein biochemistry and membrane bound enzyme alterations associated with skeletal muscles of myd/myd. 2. Our studies demonstrate that there was a progressive decline in myofibrillar ATPase activity, and this decrease is greatest in 30 weeks old animals of myd/myd as compared to controls. 3. The proteolytic activity of myofibrils isolated from myd/myd was significantly higher than controls. 4. There was no significant difference in Ca2+ ATPase activity of myosin and actin-activated myosin ATPase activity of myd/myd and their controls. 5. Mg2+ ATPase and Na(+)+K(+)-ATPase of myodystrophic SL showed significant increase compared to controls. 6. Isoproterenol stimulated adenylate cyclase activity was significantly lower in the SL of dystrophic mice compared to controls. 7. GTP+isoproterenol stimulate adenylate cyclase was significantly higher in control SL and SR when compared to SL and SR isolated from myd/myd. 8. Guanylate cyclase activity was greater in myodystrophic mice both in the absence and presence of Triton X-100. cGMP and cAMP phosphodiesterase activities were greater in dystrophic mice as compared to controls. 9. These observations suggest that there are significant changes in myofibrillar ATPase, myofibrillar protease and membrane bound enzymes of myd/myd compared to control.


Assuntos
Músculos/enzimologia , Distrofia Muscular Animal/enzimologia , Miofibrilas/enzimologia , Adenosina Trifosfatases/metabolismo , Adenilil Ciclases/metabolismo , Animais , Caseínas/metabolismo , Guanilato Ciclase/metabolismo , Membranas/enzimologia , Camundongos , Músculos/ultraestrutura , Diester Fosfórico Hidrolases/metabolismo , Sarcolema/enzimologia , Retículo Sarcoplasmático/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA