Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Clin Exp Allergy ; 43(10): 1124-33, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24074330

RESUMO

BACKGROUND: The incidence of sleep-related breathing disorders is correlated with lower and upper airway inflammatory diseases, such as asthma and allergic rhinitis. We hypothesized that corticosteroids treatment would lead to a greater reduction in disease severity in obstructive sleep apnoea syndrome (OSAS) patients with concomitant allergic rhinitis vs. non-allergic OSAS patients by reducing the level of inflammation in upper airway tissues. OBJECTIVE: This study was performed to determine whether treatment with intranasal corticosteroids could reduce upper airway inflammation and improve sleep parameters in obstructive sleep apnoea syndrome patients with or without concomitant allergic rhinitis. METHODS: Obstructive sleep apnoea syndrome patients with (n = 34) or without (n = 21) documented allergic rhinitis voluntarily enrolled in the study and were assessed at baseline and after corticosteroids treatment for 10-12 weeks. Sleep studies were performed and biopsies were obtained from the inferior turbinate, nasopharynx, and uvula. The apnoea-hypopnoea index, sleep quality, and level of daytime alertness were determined, and immunocytochemistry was used to phenotype tissue inflammation. RESULTS: Standard sleep indices improved following treatment in the entire cohort of obstructive sleep apnoea syndrome patients, with greater improvement seen in the allergic rhinitis group. Allergic rhinitis patients demonstrated significantly improved O2 saturation and a lower supine apnoea-hypopnoea index score after corticosteroid treatment; similar improvements were not seen in the non-allergic rhinitis group. Eosinophilia was detected at all three sites in the allergic rhinitis group, but not in the non-allergic rhinitis group. Following treatment, fewer eosinophils and CD4 lymphocytes were documented at all three biopsy sites in the allergic group; the reduction in inflammation was less apparent in the non-allergic rhinitis group. CONCLUSION: This study has provided important molecular and clinical evidence regarding the ability of corticosteroids to reduce upper airway inflammation and improve obstructive sleep apnoea syndrome morbidity patients with concomitant allergic rhinitis.


Assuntos
Corticosteroides/uso terapêutico , Rinite/complicações , Rinite/tratamento farmacológico , Apneia Obstrutiva do Sono/complicações , Apneia Obstrutiva do Sono/tratamento farmacológico , Administração Intranasal , Administração Tópica , Corticosteroides/administração & dosagem , Corticosteroides/efeitos adversos , Adulto , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/efeitos adversos , Anti-Inflamatórios/uso terapêutico , Biomarcadores/metabolismo , Feminino , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Rinite/metabolismo , Índice de Gravidade de Doença , Apneia Obstrutiva do Sono/metabolismo , Resultado do Tratamento
3.
Eur Respir J ; 38(1): 89-97, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20847078

RESUMO

Inflammation may contribute to upper airway pathophysiology in obstructive sleep apnoea (OSA). Our objective was to compare upper airway pro-inflammatory cytokine expression, oxidative stress and connective tissue deposition in severe (n = 25) versus mild (n = 17) OSA patients. Upper airway surgical specimens were separated by predominance of either mucosal or muscle tissue. Expression levels of interleukin (IL)-1α, IL-6, interferon-γ, RANTES (regulated on activation, normal T-cell expressed and secreted), transforming growth factor (TGF)-ß and l-selectin were measured by ribonuclease protection assay. Oxidative stress was assessed via protein carbonyl group detection by immunoblotting. Histochemistry was employed for immunolocalisation of selected cytokines and connective tissue morphometry. In the severe OSA group, expression of IL-1α, IL-6 and TGF-ß was significantly higher in mucosa-predominant tissues, whereas in muscle-predominant specimens, RANTES expression was greater in severe OSA. Increased protein carbonylation was observed in severe OSA within both mucosal and muscle compartments. Immunohistochemistry localised TGF-ß to submucosal and perimuscular inflammatory cells, while IL-6 was primarily localised to myocytes. Consistent with the pro-fibrotic cytokine profile observed in mucosa-predominant tissue, morphometric analysis revealed greater submucosal and perimuscular connective tissue in severe OSA subjects. There is increased pro-inflammatory and pro-fibrotic cytokine expression, oxidative stress, and connective tissue deposition in upper airway tissues from severe versus mild OSA patients.


Assuntos
Citocinas/biossíntese , Estresse Oxidativo , Apneia Obstrutiva do Sono/metabolismo , Apneia Obstrutiva do Sono/patologia , Adulto , Citocinas/metabolismo , Feminino , Fibrose/patologia , Regulação da Expressão Gênica , Humanos , Inflamação , Interleucina-1alfa/biossíntese , Interleucina-6/biossíntese , Masculino , Pessoa de Meia-Idade , Polissonografia/métodos , Fator de Crescimento Transformador beta/biossíntese
4.
Proc Natl Acad Sci U S A ; 105(19): 7028-33, 2008 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-18474859

RESUMO

We recently demonstrated early metabolic alterations in the dystrophin-deficient mdx heart that precede overt cardiomyopathy and may represent an early "subclinical" signature of a defective nitric oxide (NO)/cGMP pathway. In this study, we used genetic and pharmacological approaches to test the hypothesis that enhancing cGMP, downstream of NO formation, improves the contractile function, energy metabolism, and sarcolemmal integrity of the mdx heart. We first generated mdx mice overexpressing, in a cardiomyocyte-specific manner, guanylyl cyclase (GC) (mdx/GC(+/0)). When perfused ex vivo in the working mode, 12- and 20-week-old hearts maintained their contractile performance, as opposed to the severe deterioration observed in age-matched mdx hearts, which also displayed two to three times more lactate dehydrogenase release than mdx/GC(+/0). At the metabolic level, mdx/GC(+/0) displayed a pattern of substrate selection for energy production that was similar to that of their mdx counterparts, but levels of citric acid cycle intermediates were significantly higher (36 +/- 8%), suggesting improved mitochondrial function. Finally, the ability of dystrophin-deficient hearts to resist sarcolemmal damage induced in vivo by increasing the cardiac workload acutely with isoproterenol was enhanced by the presence of the transgene and even more so by inhibiting cGMP breakdown using the phosphodiesterase inhibitor sildenafil (44.4 +/- 1.0% reduction in cardiomyocyte damage). Overall, these findings demonstrate that enhancing cGMP signaling, specifically downstream and independent of NO formation, in the dystrophin-deficient heart improves contractile performance, myocardial metabolic status, and sarcolemmal integrity and thus constitutes a potential clinical avenue for the treatment of the dystrophin-related cardiomyopathies.


Assuntos
Cardiomiopatias/prevenção & controle , GMP Cíclico/metabolismo , Distrofina/deficiência , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Piperazinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sulfonas/farmacologia , Animais , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Cardiomiopatias/enzimologia , Cardiomiopatias/fisiopatologia , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Especificidade de Órgãos/efeitos dos fármacos , Purinas/farmacologia , Sarcolema/efeitos dos fármacos , Sarcolema/metabolismo , Citrato de Sildenafila
5.
Muscle Nerve ; 24(11): 1468-75, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11745948

RESUMO

In skeletal muscle fibers, nitric oxide is synthesized by neuronal nitric oxide synthase (nNOS), which normally associates with the dystrophin complex in close proximity to the sarcolemma. Many reports have documented that very low levels of nNOS protein exist in muscle fibers of Duchenne muscular dystrophy (DMD) patients. In this study we investigated the functional significance of PIN (protein inhibitor of nNOS) in targeting of nNOS to the sarcolemma and the association between nNOS and the dystrophin complex in normal and dystrophic muscle fibers. Northern blotting for PIN mRNA in normal mouse muscles and muscles of mdx mice (an animal model of DMD) revealed a significant rise in PIN mRNA in dystrophic muscles compared with normal muscles. Immunohistochemical analysis showed that, in normal mouse muscle fibers, PIN expression was localized at the sarcolemma, peripheral nuclei, and the sarcoplasm. By comparison, PIN protein in muscles from mdx mice was more concentrated around the sarcolemma and central nuclei. The presence of PIN protein expression in muscles from mdx mice was evident despite the significant reduction in nNOS and dystrophin protein expressions in these fibers. In muscle sections of DMD patients, the absence of nNOS protein expression was accompanied by maintained PIN expression. Prominent PIN expression was also detectable in macrophages infiltrating dystrophic muscle fibers both in mdx mice and DMD patients. These results suggest that PIN expression in muscles from mdx mice and DMD patients is controlled by factors different from those involved in the regulation of nNOS and dystrophin. Moreover, our results indicate that PIN is not an integral component of the dystrophin complex inside skeletal muscle fibers.


Assuntos
Proteínas de Transporte/genética , Proteínas de Drosophila , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Óxido Nítrico Sintase/genética , Animais , Proteínas de Transporte/análise , Dineínas , Distrofina/análise , Distrofina/genética , Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculo Esquelético/química , Músculo Esquelético/fisiologia , Óxido Nítrico Sintase/análise , Óxido Nítrico Sintase Tipo I , RNA Mensageiro/análise , Sarcolema/enzimologia
6.
Mol Ther ; 4(5): 499-507, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11708887

RESUMO

Duchenne muscular dystrophy (DMD) is caused by defects in the dystrophin gene. In young dystrophic mdx mice, immature regenerating myofibers represent the principal substrate for adenovirus vector (AdV)-mediated dystrophin gene transfer. However, in DMD patients immature regenerating myofibers are generally sparse. Such a situation also exists in old mdx mice, which may represent a more realistic model. Therefore, here we have used old mdx mice (of 14- to 17 months of age) to test the hypothesis that one-time administration of a myonecrotic agent can transiently re-establish a population of immature myofibers susceptible to AdV-mediated dystrophin gene transfer. This strategy led to upregulation of the coxsackie/adenovirus attachment receptor by means of induction of regenerating myofibers, significantly augmented AdV-mediated dystrophin gene expression, and enhanced force-generating capacity. In addition, it led to an increased resistance to contraction-induced injury compared with untreated controls. The latter protective effect was positively correlated with the number of dystrophin-expressing myofibers (r=0.83, P<0.05). Accordingly, the risk:benefit ratio associated with the sequential use of forced myofiber regeneration and AdV-mediated dystrophin gene transfer was favorable in old mdx mice despite advanced disease. These findings have implications for the potential applicability of AdV-mediated gene therapy to DMD and other muscle diseases in which immature regenerating myofibers are lacking.


Assuntos
Envelhecimento/fisiologia , Distrofina/genética , Distrofina/uso terapêutico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatologia , Miofibrilas/fisiologia , Regeneração , Adenoviridae/genética , Animais , Western Blotting , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Distrofina/administração & dosagem , Venenos Elapídicos/farmacologia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Imuno-Histoquímica , Contração Isométrica , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/terapia , Miofibrilas/efeitos dos fármacos , Miofibrilas/genética , Miofibrilas/patologia , Receptores Virais/biossíntese , Receptores Virais/genética , Regeneração/efeitos dos fármacos , Estresse Mecânico , Transgenes/genética , Regulação para Cima/efeitos dos fármacos
7.
Hum Gene Ther ; 12(14): 1741-55, 2001 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-11560768

RESUMO

Helper-dependent adenoviruses (HDAd) are Ad vectors lacking all or most viral genes. They hold great promise for gene therapy of diseases such as Duchenne muscular dystrophy (DMD), because they are less immunogenic than E1/E3-deleted Ad (first-generation Ad or FGAd) and can carry the full-length (Fl) dystrophin (dys) cDNA (12 kb). We have compared the transgene expression of a HDAd (HDAdCMVDysFl) and a FGAd (FGAdCMV-dys) in cell culture (HeLa, C2C12 myotubes) and in the muscle of mdx mice (the mouse model for DMD). Both vectors encoded dystrophin regulated by the same cytomegalovirus (CMV) promoter. We demonstrate that the amount of dystrophin expressed was significantly higher after gene transfer with FGAdCMV-dys compared to HDAdCMVDysFl both in vitro and in vivo. However, gene transfer with HDAdCMVDysFl in the presence of a FGAd resulted in a significant increase of dystrophin expression indicating that gene products synthesized by the FGAd increase, in trans, the amount of dystrophin produced. This enhancement occurred in cell culture and after gene transfer in the muscle of mdx mice and dystrophic golden retriever (GRMD) dogs, another animal model for DMD. The E4 region of Ad is required for the enhancement, because no increase of dystrophin expression from HDAdCMVDysFl was observed in the presence of an E1/E4-deleted Ad in vitro and in vivo. The characterization of these enhancing gene products followed by their inclusion into an HDAd may be required to produce sufficient dystrophin to mitigate the pathology of DMD by HDAd-mediated gene transfer.


Assuntos
Adenoviridae/genética , Distrofina/biossíntese , Técnicas de Transferência de Genes , Músculos/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Linhagem Celular , Células Cultivadas , Citomegalovirus/genética , DNA Complementar/metabolismo , Cães , Vetores Genéticos , Células HeLa , Humanos , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos mdx , Fenótipo , Regiões Promotoras Genéticas , Mapeamento por Restrição , Ativação Transcricional , Transgenes
9.
Physiol Genomics ; 3(3): 133-44, 2000 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-11015608

RESUMO

Duchenne muscular dystrophy (DMD) is a fatal disease caused by defects in the gene encoding dystrophin. Dystrophin is a cytoskeletal protein, which together with its associated protein complex, helps to protect the sarcolemma from mechanical stresses associated with muscle contraction. Gene therapy efforts aimed at supplying a normal dystrophin gene to DMD muscles could be hampered by host immune system recognition of dystrophin as a "foreign" protein. In contrast, a closely related protein called utrophin is not foreign to DMD patients and is able to compensate for dystrophin deficiency when overexpressed throughout development in transgenic mice. However, the issue of which of the two candidate molecules is superior for DMD therapy has remained an open question. In this study, dystrophin and utrophin gene transfer effects on dystrophic muscle function were directly compared in the murine (mdx) model of DMD using E1/E3-deleted adenovirus vectors containing either a dystrophin (AdV-Dys) or a utrophin (AdV-Utr) transgene. In immunologically immature neonatal animals, AdV-Dys and AdV-Utr improved tibialis anterior muscle histopathology, force-generating capacity, and the ability to resist injury caused by high-stress contractions to an equivalent degree. By contrast, only AdV-Utr was able to achieve significant improvement in force generation and the ability to resist stress-induced injury in the soleus muscle of immunocompetent mature mdx animals. In addition, in mature mdx mice, there was significantly greater transgene persistence and reduced inflammation with utrophin compared to dystrophin gene transfer. We conclude that dystrophin and utrophin are largely equivalent in their intrinsic abilities to prevent the development of muscle necrosis and weakness when expressed in neonatal mdx animals with an immature immune system. However, because immunity against dystrophin places an important limitation on the efficacy of dystrophin gene replacement in an immunocompetent mature host, the use of utrophin as an alternative to dystrophin gene transfer in this setting appears to offer a significant therapeutic advantage.


Assuntos
Proteínas do Citoesqueleto/genética , Distrofina/genética , Proteínas de Membrana/genética , Distrofia Muscular de Duchenne/terapia , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/fisiologia , DNA Recombinante/genética , Distrofina/análise , Distrofina/fisiologia , Técnicas de Transferência de Genes , Terapia Genética , Membro Posterior , Imunocompetência , Imuno-Histoquímica , Inflamação/genética , Inflamação/patologia , Proteínas de Membrana/análise , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos mdx , Contração Muscular , Músculos/química , Músculos/metabolismo , Músculos/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatologia , Estresse Mecânico , Utrofina
10.
Hum Gene Ther ; 11(5): 701-14, 2000 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-10757350

RESUMO

Duchenne muscular dystrophy (DMD) and other inherited myopathies lead to progressive destruction of most skeletal muscles in the body, including those responsible for maintaining respiration. DMD is a fatal disorder caused by defects in the dystrophin gene. Recombinant adenovirus vectors (AdV) are considered a promising means for therapeutic delivery of a functional dystrophin gene to DMD muscles. If AdV-mediated dystrophin gene replacement in DMD is to be successful, development of a systemic delivery method for targeting the large number of diseased muscles will be required. In this study we investigated two major factors preventing efficient AdV-mediated gene transfer to skeletal muscles of adult animals after intravascular AdV administration: (1) an inability of AdV particles to breach the endothelial barrier and enter into contact with myofibers, and (2) a relatively nonpermissive myofiber population for AdV infection due at least in part to insufficient levels of the coxsackie/adenovirus attachment receptor (CAR). On the basis of established principles governing the transendothelial flux of macromolecules, we further hypothesized that an alteration in Starling forces (increased hydrostatic and decreased osmotic pressures) within the intravascular compartment would facilitate AdV transendothelial flux via convective transport. In addition, experimental muscle regeneration was employed to increase the prevalence of immature myofibers in which CAR expression is upregulated. Here we report that by employing the above-described strategy, high-level heterologous reporter gene expression was achievable in hindlimb muscles of normal rats as well as dystrophic (mdx) mice (genetic homolog of DMD) after a single intraarterial injection of AdV. Microsphere studies confirmed enhanced transport into muscle of fluorescent tracer particles in the size range of AdV, and there was a high concordance between CAR upregulation and myofiber transduction after intraarterial AdV delivery. Furthermore, in mdx mice examined 10 days after intraarterial AdV delivery, the aforementioned procedures had no adverse effects on the force-generating capacity of targeted muscles. These findings have implications for eventual AdV-mediated gene therapy of generalized skeletal muscle diseases such as DMD using a systemic intraarterial delivery approach.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/patologia , Animais , Fenômenos Biomecânicos , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Distrofina/genética , Regulação da Expressão Gênica , Injeções Intra-Arteriais , Camundongos , Camundongos Mutantes , Fibras Musculares Esqueléticas/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Distrofias Musculares/genética , Distrofias Musculares/patologia , Distrofias Musculares/terapia , Ratos , Ratos Sprague-Dawley , Receptores Virais/genética , Receptores Virais/metabolismo
11.
Hum Gene Ther ; 10(8): 1299-310, 1999 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-10365661

RESUMO

Utrophin is a close homolog of dystrophin, the protein whose mutations cause Duchenne muscular dystrophy (DMD). Utrophin is present at low levels in normal and dystrophic muscle, whereas dystrophin is largely absent in DMD. In such cases, the replacement of dystrophin using a utrophin gene transfer strategy could be more advantageous because utrophin would not be a neoantigen. To establish if adenovirus (AV)-mediated utrophin gene transfer is a possible option for the treatment of DMD, an AV vector expressing a shortened version of utrophin (AdCMV-Utr) was constructed. The effect of utrophin overexpression was investigated following intramuscular injection of this AV into mdx mice, the mouse model of DMD. When the tibialis anterior (TA) muscles of 3- to 5-day-old animals were injected with 5 microl of AdCMV-Utr (7.0 x 10(11) virus/ml), an average of 32% of fibers were transduced and the transduction level remained stable for at least 60 days. The presence of utrophin restored the normal histochemical pattern of the dystrophin-associated protein complex at the cell surface and resulted in a reduction in the number of centrally nucleated fibers. The transduced fibers were largely impermeable to the tracer dye Evans blue, suggesting that utrophin protects the surface membrane from breakage. In vitro measurements of the force decline in response to high-stress eccentric contractions demonstrated that the muscles overexpressing utrophin were more resistant to mechanical stress-induced injury. Taken together, these data indicate that AV-mediated utrophin gene transfer can correct various aspects of the dystrophic phenotype. However, a progressive reduction in the number of transduced fibers was observed when the TA muscles of 30- to 45-day-old mice were injected with 25 microl of AdCMV-Utr. This reduction coincides with a humoral response to the AV and transgene, which consists of a hybrid mouse-human cDNA.


Assuntos
Adenoviridae , Proteínas do Citoesqueleto/uso terapêutico , Técnicas de Transferência de Genes , Vetores Genéticos , Proteínas de Membrana/uso terapêutico , Distrofias Musculares/terapia , Animais , Animais Recém-Nascidos , Proteínas do Citoesqueleto/genética , Expressão Gênica , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos mdx , Músculos/patologia , Distrofias Musculares/patologia , Fenótipo , Utrofina
12.
Am J Respir Cell Mol Biol ; 20(2): 319-26, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9922224

RESUMO

In skeletal muscle fibers, nitric oxide (NO) is synthesized by neuronal NO synthase (nNOS) and regulates excitation-contraction coupling, glucose uptake, and mitochondrial respiration. Recently, a novel 89-amino acid protein, designated protein inhibitor of nNOS (PIN), has been shown to interact with and specifically inhibit nNOS activity. In this study, we investigated the distribution, localization, and regulation of PIN expression in ventilatory and limb muscles of various species. Amplified PIN cDNA from the rat diaphragm revealed an open reading frame identical to that of human PIN. Among muscles of adult rats, PIN mRNA was strongly expressed in muscles rich in type I fibers, whereas much weaker expression was evident in muscles rich in type II fibers. By comparison, PIN protein expression was not related to fiber-type distribution. Similarly, PIN protein was equally expressed among rat, mouse, and human diaphragms. Both PIN mRNA and PIN protein were expressed at much higher levels in the embryonic rat diaphragm than in adult muscle. Immunohistochemistry revealed that PIN protein was localized in close proximity to the sarcolemma and nuclei. PIN protein was also abundant in muscle spindles and axons of nerves supplying skeletal muscle fibers. We conclude that PIN is expressed in various skeletal muscle fibers and that its expression is regulated during muscle development. The localization of PIN in muscle regions containing abundant nNOS protein suggests that it plays a role in the regulation of NO synthesis in skeletal muscle fibers.


Assuntos
Proteínas de Transporte/metabolismo , Diafragma/metabolismo , Proteínas de Drosophila , Óxido Nítrico Sintase/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Transporte/genética , Clonagem Molecular , DNA Complementar , Diafragma/enzimologia , Dineínas , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo , Óxido Nítrico Sintase Tipo I , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie
13.
Am J Respir Crit Care Med ; 158(5 Pt 1): 1656-63, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9817721

RESUMO

Endotoxemia is associated with impaired diaphragm contractility, and increased nitric oxide (NO) production has recently been implicated in this phenomenon. However, the precise nature of sepsis-related alterations in diaphragm myofiber function remains unclear. We tested the hypothesis that enhanced NO synthesis during sepsis produces diaphragm sarcolemmal injury with attendant abnormalities of myofiber membrane electrophysiology. Two different rat sepsis models were employed: acute (4 h) intraarterial endotoxin (LPS; 20 mg/kg) and subacute (24 h) peritonitis induced by cecal ligation and perforation (CLP). Diaphragm damage occurred after both LPS and CLP, as indicated by hyperpermeability of myofibers to a low molecular weight tracer dye, which is normally unable to penetrate the sarcolemma. Sarcolemmal injury was significantly correlated with reductions in the resting membrane potential (Em) of single diaphragm myofibers. Western analysis revealed increased diaphragmatic expression of the inducible isoform of NO synthase (iNOS) after LPS and CLP. An inhibitor of NOS activity, LNMMA, significantly decreased morphologic as well as electrophysiologic signs of myofiber membrane injury and dysfunction. Therefore, we conclude that both acute endotoxemia and subacute peritonitis models of sepsis lead to significant sarcolemmal damage and altered Em in diaphragm myofibers. These changes appear to be mediated, at least in part, through the pathway of increased nitric oxide production.


Assuntos
Diafragma/patologia , Inibidores Enzimáticos/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Sarcolema/patologia , Sepse/patologia , ômega-N-Metilarginina/farmacologia , Animais , Western Blotting , Doenças do Ceco/complicações , Permeabilidade da Membrana Celular , Corantes , Diafragma/efeitos dos fármacos , Modelos Animais de Doenças , Endotoxinas/efeitos adversos , Escherichia coli , Perfuração Intestinal/complicações , Lipopolissacarídeos/efeitos adversos , Masculino , Potenciais da Membrana/fisiologia , Contração Muscular , Fibras Musculares Esqueléticas/patologia , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/biossíntese , Peritonite/etiologia , Ratos , Ratos Sprague-Dawley , Sarcolema/efeitos dos fármacos , Sepse/enzimologia
14.
Am J Physiol ; 274(6): L980-9, 1998 06.
Artigo em Inglês | MEDLINE | ID: mdl-9609737

RESUMO

Although prolonged diaphragm denervation (DNV) produces myofiber atrophy and a loss of type I myosin heavy chain (MHC) expression, short-term DNV leads to significant diaphragm hypertrophy. The purpose of this study was to explore the regulation of MHC isoform expression and muscle remodeling during DNV hypertrophy of the diaphragm. Both unilateral and bilateral DNV led to similar changes, with a significant increase in total RNA content and muscle mass but no change in dry-to-wet weight ratio. Sarcomere number was also increased in diaphragm myofibers after DNV ( approximately 20%), suggesting an adaptive response to muscle stretch. There was hypertrophy of type I myofibers and increased coexpression of type I and type II MHCs within single myofibers by immunocytochemistry as well as increased type I MHC (25-46%) and decreased type IIb MHC (14-39%) by SDS-PAGE. Contractility parameters were also consistent with a type II-to-type I MHC phenotype transformation. Importantly, DNV-induced modulation of MHC isoform mRNA transcript levels did not correspond to changes in their cognate proteins, suggesting a major degree of posttranscriptional control. We conclude that DNV hypertrophy of the diaphragm is associated with reciprocal changes in type I and type II MHC isoforms that are directly opposed to the type I-to-type II MHC phenotype transformation reported in the diaphragm DNV atrophy model. Furthermore, in contradistinction to most hypertrophy models, control of MHC gene expression and myofibrillar remodeling after short-term DNV appears to entail major involvement of posttranscriptional regulatory mechanisms.


Assuntos
Diafragma/inervação , Regulação da Expressão Gênica , Denervação Muscular , Cadeias Pesadas de Miosina/genética , Animais , Northern Blotting , Diafragma/patologia , Diafragma/fisiologia , Eletroforese em Gel de Poliacrilamida , Hipertrofia , Imuno-Histoquímica , Masculino , Contração Muscular , Cadeias Pesadas de Miosina/isolamento & purificação , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Sarcômeros/patologia
15.
Mol Cell Biochem ; 179(1-2): 111-23, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9543354

RESUMO

Duchenne muscular dystrophy (DMD) is the most common of the human muscular dystrophies, affecting approximately 1 in 3500 boys. Most DMD patients die in their late teens or early twenties due to involvement of the diaphragm and other respiratory muscles by the disease. The primary abnormality in DMD is an absence of dystrophin, a 427 kd protein normally found at the cytoplasmic face of the muscle cell surface membrane. Based upon the predicted structure and location of the protein, it has been proposed that dystrophin plays an important role in providing mechanical reinforcement to the sarcolemmal membrane of muscle fibers. Therefore, dystrophin could help to protect muscle fibers from potentially damaging tissue stresses developed during muscle contraction. In the present paper, the nature of mechanical stresses placed upon myofibers during various forms of muscle contraction are reviewed, along with current lines of evidence supporting a critical role for dystrophin as a subsarcolemmal membrane-stabilizing protein in this setting. In addition, the implications of these findings for exercise programs and other potential forms of therapy in DMD are discussed.


Assuntos
Distrofias Musculares/genética , Músculos Respiratórios/patologia , Animais , Modelos Animais de Doenças , Distrofina/deficiência , Distrofina/genética , Exercício Físico/fisiologia , Histocitoquímica , Humanos , Camundongos , Camundongos Endogâmicos mdx , Modelos Moleculares , Contração Muscular/fisiologia , Proteínas Musculares/química , Distrofias Musculares/terapia
16.
Gene Ther ; 5(3): 369-79, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9614557

RESUMO

Duchenne muscular dystrophy (DMD) and murine X-linked muscular dystrophy (mdx) are both due to absence of the subsarcolemmal protein dystrophin. Recombinant adenovirus vectors (AdV) are considered a promising means for delivering a functional dystrophin gene to muscle. However, the usefulness of AdV for this purpose is limited by vector toxicity as well as immune-mediated elimination of infected fibers. In addition, studies to date of AdV-mediated dystrophin gene transfer have either failed to examine effects on muscle strength or been performed in immunologically immature neonatal animals with little baseline abnormality of force-generating capacity. In the present study, AdV-mediated dystrophin gene transfer was performed in adult mdx mice with pre-existent dystrophic pathology and muscle weakness. The main findings are as follows: (1) acute myofiber toxicity and gene transfer efficiency are both AdV dose-dependent, such that the therapeutic margin of safety is fairly narrow; (2) immunosuppressive therapy (FK506) prevents immune-mediated elimination of dystrophin-positive fibers but not the dose-dependent toxic effects; (3) at the optimal vector dosage and with effective immunosuppression, AdV-mediated dystrophin minigene transfer is capable of alleviating the loss of force-generating capacity as well as histopathological evidence of disease progression normally seen in adult mdx muscles over a 2-month period. These findings have important implications for the eventual application of AdV-mediated dystrophin gene transfer in DMD patients.


Assuntos
Adenoviridae/genética , Distrofina/genética , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Distrofias Musculares/terapia , Animais , Diafragma/patologia , Diafragma/fisiopatologia , Distrofina/metabolismo , Técnicas de Transferência de Genes , Terapia Genética/efeitos adversos , Terapia de Imunossupressão , Masculino , Camundongos , Camundongos Endogâmicos mdx , Contração Muscular , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofias Musculares/patologia , Distrofias Musculares/fisiopatologia
17.
J Virol ; 72(6): 4601-9, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9573223

RESUMO

Recombinant adenovirus vectors (AdV) have been considered a potential vehicle for performing gene therapy in patients suffering from Duchenne muscular dystrophy but are limited by a cellular and humoral immune response that prevents long-term transgene expression as well as effective transduction after AdV readministration. Conventional immunosuppressive agents such as cyclosporine and FK506, which act by interfering with CD3-T-cell receptor-mediated signaling via calcineurin, are only partially effective in reversing these phenomena and may also produce substantial organ toxicity. We hypothesized that activation of redundant T-cell activation pathways could limit the effectiveness of these drugs at clinically tolerable doses. Therefore, we have tested the ability of immunomodulatory immunoglobulins (Ig) with different modes of action to facilitate AdV-mediated gene transfer to adult dystrophic (mdx) mice. When used in isolation, immunomodulatory Ig (anti-intercellular adhesion molecule-1, anti-leukocyte function-associated antigen-1, anti-CD2, and CTLA4Ig) were only mildly effective in mitigating cellular and/or humoral immunity against adenovirus capsid proteins and the therapeutic transgene product, dystrophin. However, the combination of FK506 plus CTLA4Ig abrogated the immune response against adenovirus proteins and dystrophin to a degree not achievable with the use of either agent alone. At 30 days after AdV injection, >90% of myofibers could be found to express dystrophin with little or no evidence of a cellular immune response against transduced fibers. In addition, the humoral immune response was markedly suppressed, and this was associated with increased transduction efficiency following vector readministration. These data suggest that by facilitating both primary and secondary transduction after AdV administration, combined targeting of CD3-T-cell receptor-mediated signaling via calcineurin and the B7:CD28 costimulatory pathway could greatly increase the potential utility of AdV-mediated gene transfer as a therapeutic modality for genetic diseases such as Duchenne muscular dystrophy that will require long-term transgene expression and repeated vector delivery.


Assuntos
Adenoviridae , Adjuvantes Imunológicos/farmacologia , Antígenos CD28/imunologia , Calcineurina/imunologia , Técnicas de Transferência de Genes , Vetores Genéticos , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/terapia , Transdução de Sinais/imunologia , Adenoviridae/imunologia , Animais , Antígenos CD28/genética , Calcineurina/genética , Distrofina/imunologia , Terapia Genética , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Distrofia Muscular Animal/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Linfócitos T/imunologia
18.
Eur Respir J ; 11(2): 492-7, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9551759

RESUMO

The protein dystrophin is absent in the muscles of patients with Duchenne muscular dystrophy (DMD) as well as dystrophin-deficient mice with muscular dystrophy (mdx mice). The mdx mouse diaphragm closely resembles the human DMD phenotype and thus provides a useful model for studies of dystrophin gene replacement. Recombinant adenovirus vectors (AdVs) hold promise as a means for delivering a functional dystrophin gene to muscle. As an initial step toward this goal, we have determined the efficiency and functional consequences of AdV-mediated reporter gene transfer to the diaphragm in both normal and mdx adult mice. At 1 week after AdV administration, there was a high level of transgene expression in the diaphragm. One month later, however, elimination of transgene expression was observed along with a significant decrease in force production by both normal and mdx diaphragms. Immunosuppression with cyclosporine did not augment the level of transgene expression, but a beneficial effect on diaphragm force-generating capacity was observed in both groups of animals. In order to further elucidate the cellular mechanisms underlying these findings, the effects of AdV gene inactivation (by ultraviolet (UV) irradiation) and interference with host T-lymphocyte subsets were examined. Both UV-inactivation of AdV and CD8+ T-cell deficiency were found to significantly alleviate AdV-induced reductions in diaphragm force-generating capacity. Brief (2 day) administration of a neutralizing antibody against host CD4+ T-cells also produced a trend towards mitigation of AdV-induced contractile dysfunction. In addition, transgene expression one month after AdV delivery was significantly enhanced with inhibition of either CD4+ or CD8+ T-cell function. The data suggest two major sources of reduced force generation after recombinant adenovirus vector-mediated gene transfer to muscle: 1) a cytotoxic component associated with recombinant adenovirus vector transcriptional activity; and 2) an immune-based component of more delayed onset that is primarily dependent upon CD8+ T-cell activity. These results have important implications for the design of future generation vectors and the potential need for immunosuppressive therapy after recombinant adenovirus vector mediated dystrophin gene transfer to Duchenne muscular dystrophy patients.


Assuntos
Adenoviridae/genética , Marcação de Genes , Terapia Genética , Vetores Genéticos , Distrofias Musculares/terapia , Músculos Respiratórios/fisiopatologia , Animais , Humanos
19.
Muscle Nerve ; 21(3): 394-7, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9486869

RESUMO

The effects of methylprednisolone therapy on the susceptibility of dystrophin-deficient myofibers to contraction-induced injury were evaluated in the mdx mouse diaphragm model of Duchenne dystrophy. Mdx myofibers were abnormally vulnerable to injury induced by high-stress eccentric contractions. However, methylprednisolone therapy did not significantly alter the degree of contraction-induced injury. These data suggest that beneficial effects of corticosteroid therapy in Duchenne dystrophy are unlikely to be related to a change in the threshold for contraction-induced myofiber damage.


Assuntos
Corticosteroides/uso terapêutico , Metilprednisolona/uso terapêutico , Contração Muscular/fisiologia , Músculo Esquelético/lesões , Distrofia Muscular Animal/tratamento farmacológico , Distrofia Muscular Animal/fisiopatologia , Análise de Variância , Animais , Diafragma , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia
20.
Curr Opin Neurol ; 10(5): 430-5, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9330891

RESUMO

Gene therapy is a promising option for the definitive treatment of Duchenne and Becker muscular dystrophies. Presently, gene therapy for Duchenne and Becker muscular dystrophies is still in the preclinical stage with dystrophin-deficient animals (the mdx mouse and a golden retriever dog strain) serving as convenient models. The thrust of research during the past 18 months has focused on two approaches: adenovirus-mediated dystrophin gene transfer and upregulation of a natural dystrophin analogue, utrophin. In the area of adenovirus-mediated gene transfer, substantial progress has been made in characterizing and mitigating the deleterious immune responses to the vector and transgene proteins. Furthermore, new adenovirus vectors have been created with reduced immunogenicity and increased insert gene capacity, which enhance the longevity of the transgene expression. Additional efforts are underway to develop safe and efficient routes of administration of the adenovirus vector carrying the dystrophin expression cassette. The prospects of utrophin upregulation as an attractive strategy for treatment of Duchenne and Becker muscular dystrophies was greatly enhanced by the demonstration of a substantial mitigation of the dystrophic phenotype of the transgenic mdx mouse overexpressing utrophin.


Assuntos
Terapia Genética , Distrofias Musculares/terapia , Animais , Proteínas do Citoesqueleto/genética , Cães , Distrofina/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos mdx , Distrofias Musculares/genética , Distrofia Muscular Animal/genética , Pesquisa , Utrofina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...