Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-32998454

RESUMO

BACKGROUND: We are developing a novel therapy for Duchenne muscular dystrophy (DMD), involving the transplantation of autologous, skeletal muscle-derived stem cells that have been genetically corrected to express dystrophin. Dystrophin is normally expressed in activated satellite cells and in differentiated muscle fibres. However, in past preclinical validation studies, dystrophin transgenes have generally been driven by constitutive promoters that would be active at every stage of the myogenic differentiation process, including in proliferating muscle stem cells. It is not known whether artificial dystrophin expression would affect the properties of these cells. AIMS: Our aims are to determine if mini-dystrophin expression affects the proliferation or myogenic differentiation of DMD skeletal muscle-derived cells. METHODS: Skeletal muscle-derived cells from a DMD patient were transduced with lentivirus coding for mini-dystrophins (R3-R13 spectrin-like repeats (ΔR3R13) or hinge2 to spectrin-like repeats R23 (ΔH2R23)) with EGFP (enhanced green fluorescence protein) fused to the C-terminus, driven by a constitutive promoter, spleen focus-forming virus (SFFV). Transduced cells were purified on the basis of GFP expression. Their proliferation and myogenic differentiation were quantified by ethynyl deoxyuridine (EdU) incorporation and fusion index. Furthermore, dystrophin small interfering ribonucleic acids (siRNAs) were transfected to the cells to reverse the effects of the mini-dystrophin. Finally, a phospho-mitogen-activated protein kinase (MAPK) array assay was performed to investigate signalling pathway changes caused by dystrophin expression. RESULTS: Cell proliferation was not affected in cells transduced with ΔR3R13, but was significantly increased in cells transduced with ΔH2R23. The fusion index of myotubes derived from both ΔR3R13- and ΔH2R23 -expressing cells was significantly compromised in comparison to myotubes derived from non-transduced cells. Dystrophin siRNA transfection restored the differentiation of ΔH2R23-expressing cells. The Erk1/2- signalling pathway is altered in cells transduced with mini-dystrophin constructs. CONCLUSIONS: Ectopic expression of dystrophin in cultured human skeletal muscle-derived cells may affect their proliferation and differentiation capacity. Caution should be taken when considering genetic correction of autologous stem cells to express dystrophin driven by a constitutive promoter.


Assuntos
Distrofina/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Transgenes , Diferenciação Celular , Engenharia Celular/métodos , Proliferação de Células , Distrofina/antagonistas & inibidores , Distrofina/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Sistema de Sinalização das MAP Quinases , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Plasmídeos/química , Plasmídeos/metabolismo , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Espectrina/genética , Espectrina/metabolismo , Transdução Genética
3.
Nucleic Acid Ther ; 29(2): 92-103, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30672725

RESUMO

Duchenne muscular dystrophy is a severe, progressive muscle-wasting disease that is caused by mutations that abolish the production of functional dystrophin protein. The exon skipping approach aims to restore the disrupted dystrophin reading frame, to allow the production of partially functional dystrophins, such as found in the less severe Becker muscular dystrophy. Exon skipping is achieved by antisense oligonucleotides (AONs). Several chemical modifications have been tested in nonclinical and clinical trials. The morpholino phosphorodiamidate oligomer eteplirsen has been approved by the Food and Drug Administration, whereas clinical development with the 2'-O-methyl phosphorothioate (2OMePS) AON drisapersen was recently stopped. In this study, we aimed to study various aspects of 2OMePS AONs in nonclinical animal studies. We show that while efficiency of exon skipping restoration is comparable in young and older C57BL/10ScSn-Dmdmdx/J (mdx/BL10) mice, functional improvement was only observed for younger treated mice. Muscle quality did not affect exon skipping efficiency as exon skip and dystrophin levels were similar between mdx/BL10 and more severely affected, age-matched D2-mdx mice. We further report that treadmill running increases AON uptake and dystrophin levels in mdx/BL10 mice. Finally, we show that even low levels of exon skipping and dystrophin restoration are sufficient to significantly increase the survival of mdx-utrn-/- mice from 70 to 97 days.


Assuntos
Distrofina/genética , Distrofia Muscular de Duchenne/genética , Oligonucleotídeos Antissenso/farmacologia , Utrofina/genética , Animais , Ensaios Clínicos como Assunto , Distrofina/antagonistas & inibidores , Éxons/efeitos dos fármacos , Éxons/genética , Terapia Genética , Humanos , Camundongos , Camundongos Endogâmicos mdx , Morfolinos/genética , Morfolinos/farmacologia , Atrofia Muscular Espinal , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos/farmacologia , Oligonucleotídeos Antissenso/genética , Compostos Organotiofosforados/farmacologia
4.
J Mol Biol ; 431(9): 1780-1791, 2019 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-30597161

RESUMO

RNA is accurately entangled in virtually all pathways that maintain cellular homeostasis. To name but a few, RNA is the "messenger" between DNA encoded information and the resulting proteins. Furthermore, RNAs regulate diverse processes by forming DNA::RNA or RNA::RNA interactions. Finally, RNA itself can be the scaffold for ribonucleoprotein complexes, for example, ribosomes or cellular bodies. Consequently, disruption of any of these processes can lead to disease. This review describes known and emerging RNA-based disease mechanisms like interference with regular splicing, the anomalous appearance of RNA-protein complexes and uncommon RNA species, as well as non-canonical translation. Due to the complexity and entanglement of the above-mentioned pathways, only few drugs are available that target RNA-based disease mechanisms. However, advances in our understanding how RNA is involved in and modulates cellular homeostasis might pave the way to novel treatments.


Assuntos
DNA/genética , Terapia de Alvo Molecular/métodos , Doenças Neurodegenerativas/genética , Splicing de RNA , RNA/genética , Animais , DNA/metabolismo , Modelos Animais de Doenças , Distrofina/antagonistas & inibidores , Distrofina/genética , Distrofina/metabolismo , Humanos , Lamina Tipo A/antagonistas & inibidores , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/terapia , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Biossíntese de Proteínas , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , RNA/classificação , RNA/metabolismo , Interferência de RNA , Transcrição Gênica , Proteínas tau/antagonistas & inibidores , Proteínas tau/genética , Proteínas tau/metabolismo
5.
Hum Mutat ; 39(9): 1193-1202, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29907980

RESUMO

Antisense oligonucleotide (AON)-mediated exon skipping is an emerging therapeutic for individuals with Duchenne muscular dystrophy (DMD). Skipping of exons adjacent to common exon deletions in DMD using AONs can produce in-frame transcripts and functional protein. Targeted skipping of DMD exons 8, 44, 45, 50, 51, 52, 53, and 55 is predicted to benefit 47% of affected individuals. We observed a correlation between mutation subgroups and age at loss of ambulation in the Duchenne Registry, a large database of phenotypic and genetic data for DMD (N = 765). Males amenable to exon 44 (N = 74) and exon 8 skipping (N = 18) showed prolonged ambulation compared to other exon skip groups and nonsense mutations (P = 0.035 and P < 0.01, respectively). In particular, exon 45 deletions were associated with prolonged age at loss of ambulation relative to the rest of the exon 44 skip amenable cohort and other DMD mutations. Exon 3-7 deletions also showed prolonged ambulation relative to all other exon 8 skippable mutations. Cultured myotubes from DMD patients with deletions of exons 3-7 or exon 45 showed higher endogenous skipping than other mutations, providing a potential biological rationale for our observations. These results highlight the utility of aggregating phenotypic and genotypic data for rare pediatric diseases to reveal progression differences, identify potentially confounding factors, and probe molecular mechanisms that may affect disease severity.


Assuntos
Distrofina/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Oligodesoxirribonucleotídeos Antissenso/genética , Adolescente , Adulto , Fatores Etários , Biópsia , Códon sem Sentido/genética , Distrofina/antagonistas & inibidores , Éxons/genética , Feminino , Fibroblastos/patologia , Genótipo , Humanos , Estimativa de Kaplan-Meier , Tempo de Internação , Masculino , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/terapia , Mioblastos/patologia , Oligodesoxirribonucleotídeos Antissenso/uso terapêutico , Cultura Primária de Células , Sistema de Registros , Deleção de Sequência/genética , Caracteres Sexuais , Adulto Jovem
6.
Exp Cell Res ; 364(2): 224-233, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29458173

RESUMO

Duchenne muscular dystrophy (DMD) is an X chromosome-linked disorder caused by a mutation in the dystrophin gene. Many previous studies reported that the skeletal muscles of DMD patients were more susceptible to oxidative stress than those of healthy people. However, not much has been known about the responsible mechanism of the differential susceptibility. In this study, we established dystrophin knock-down (DysKD) cell lines by transfection of dystrophin shRNA lentiviral particles into C2 cells and found that DysKD myotubes are more vulnerable to menadione-induced oxidative stress than control myotubes. We focused on the nuclear erythroid 2-related factor 2 (Nrf2) which is a transcription factor that regulates the expression of phase II antioxidant enzymes by binding to the antioxidant response element (ARE). Under menadione-induced oxidative stress, the translocation of Nrf2 to the nucleus is significantly decreased in the DysKD myotubes. In addition, the binding of Nrf2 to ARE site of Bcl-2 gene as well as protein expression of Bcl-2 is decreased compared to the control cells. Interestingly, sulforaphane increased Akt activation and Nrf2 translocation to the nucleus in the DysKD myotubes. These results suggest that the Nrf2 pathway might be the responsible pathway to the oxidative stress-induced muscle damage in DMD.


Assuntos
Elementos de Resposta Antioxidante/efeitos dos fármacos , Distrofina/antagonistas & inibidores , Fibras Musculares Esqueléticas/efeitos dos fármacos , Distrofia Muscular de Duchenne/tratamento farmacológico , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais , Vitamina K 3/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Distrofina/metabolismo , Humanos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
Methods Mol Biol ; 1687: 123-141, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29067660

RESUMO

Exon skipping therapy using synthetic DNA-like molecules called antisense oligonucleotides (ASOs) is a promising therapeutic candidate for overcoming the dystrophin mutation that causes Duchenne muscular dystrophy (DMD). This treatment involves splicing out the frame-disrupting segment of the dystrophin mRNA, which restores the reading frame and produces a truncated yet functional dystrophin protein. Phosphorodiamidate morpholino oligomer (PMO) is the safest ASO for patients among ASOs and has recently been approved under the accelerated approval pathway by the U.S. Food and Drug Administration (FDA) as the first drug for DMD. Here, we describe the methodology and protocol of PMO transfection and evaluation of the exon skipping efficacy in the mdx52 mouse, an exon 52 deletion model of DMD produced by gene targeting. The mdx52 mouse model offers advantages over the mdx mouse, a spontaneous DMD model with a nonsense mutation in exon 23, in terms of the deletion in a hotspot of deletion mutations in DMD patients, the analysis of caveolae and also Dp140 and Dp260, shorter dystrophin isoforms.


Assuntos
Distrofina/genética , Terapia Genética/métodos , Morfolinos/uso terapêutico , Distrofia Muscular de Duchenne/terapia , Processamento Alternativo/genética , Animais , Modelos Animais de Doenças , Distrofina/antagonistas & inibidores , Éxons/genética , Humanos , Camundongos , Camundongos Endogâmicos mdx , Morfolinos/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/uso terapêutico , Transfecção
8.
Cell Death Differ ; 24(2): 330-342, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27834955

RESUMO

Dystrophin deficiency is the genetic basis for Duchenne muscular dystrophy (DMD), but the cellular basis of progressive myofiber death in DMD is not fully understood. Using two dystrophin-deficient mdx mouse models, we find that the mitochondrial dysfunction is among the earliest cellular deficits of mdx muscles. Mitochondria in dystrophic myofibers also respond poorly to sarcolemmal injury. These mitochondrial deficits reduce the ability of dystrophic muscle cell membranes to repair and are associated with a compensatory increase in dysferlin-mediated membrane repair proteins. Dysferlin deficit in mdx mice further compromises myofiber cell membrane repair and enhances the muscle pathology at an asymptomatic age for dysferlin-deficient mice. Restoring partial dystrophin expression by exon skipping improves mitochondrial function and offers potential to improve myofiber repair. These findings identify that mitochondrial deficit in muscular dystrophy compromises the repair of injured myofibers and show that this repair mechanism is distinct from and complimentary to the dysferlin-mediated repair of injured myofibers.


Assuntos
Membrana Celular/metabolismo , Mitocôndrias/metabolismo , Animais , Linhagem Celular , Disferlina/deficiência , Disferlina/genética , Distrofina/antagonistas & inibidores , Distrofina/genética , Distrofina/metabolismo , Transferência Ressonante de Energia de Fluorescência , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Microscopia de Fluorescência , Mitocôndrias/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Contração Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patologia , Mioblastos/citologia , Mioblastos/metabolismo , Oligodesoxirribonucleotídeos Antissenso/metabolismo , Ácido Pirúvico/farmacologia , Imagem com Lapso de Tempo
9.
Proc Natl Acad Sci U S A ; 111(28): E2905-14, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24969422

RESUMO

Duchenne muscular dystrophy is a fatal X-linked disease characterized by the absence of dystrophin. Approximately 20% of boys will die of dilated cardiomyopathy that is associated with cytoskeletal protein disarray, contractile dysfunction, and reduced energy production. However, the mechanisms for altered energy metabolism are not yet fully clarified. Calcium influx through the L-type Ca(2+) channel is critical for maintaining cardiac excitation and contraction. The L-type Ca(2+) channel also regulates mitochondrial function and metabolic activity via transmission of movement of the auxiliary beta subunit through intermediate filament proteins. Here, we find that activation of the L-type Ca(2+) channel is unable to induce increases in mitochondrial membrane potential and metabolic activity in intact cardiac myocytes from the murine model of Duchenne muscular dystrophy (mdx) despite robust increases recorded in wt myocytes. Treatment of mdx mice with morpholino oligomers to induce exon skipping of dystrophin exon 23 (that results in functional dystrophin accumulation) or application of a peptide that resulted in block of voltage-dependent anion channel (VDAC) "rescued" mitochondrial membrane potential and metabolic activity in mdx myocytes. The mitochondrial VDAC coimmunoprecipitated with the L-type Ca(2+) channel. We conclude that the absence of dystrophin in the mdx ventricular myocyte leads to impaired functional communication between the L-type Ca(2+) channel and mitochondrial VDAC. This appears to contribute to metabolic inhibition. These findings provide new mechanistic and functional insight into cardiomyopathy associated with Duchenne muscular dystrophy.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Distrofina/metabolismo , Potencial da Membrana Mitocondrial , Membranas Mitocondriais/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Cálcio/metabolismo , Distrofina/antagonistas & inibidores , Distrofina/genética , Éxons , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Masculino , Camundongos , Camundongos Endogâmicos mdx , Membranas Mitocondriais/patologia , Morfolinos/genética , Morfolinos/farmacologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Miocárdio , Miócitos Cardíacos/patologia
10.
Mol Pharm ; 11(3): 1053-61, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-24433046

RESUMO

Duchenne muscular dystrophy (DMD) is a genetic disorder that is caused by mutations in the DMD gene that lead to an absence of functional protein. The mdx dystrophic mouse contains a nonsense mutation in exon 23 of the dystrophin gene; a phosphorodiamidate morpholino oligomer (PMO) designed to skip this mutated exon in the mRNA induces dystrophin expression. However, an efficient PMO delivery method is needed to improve treatment strategies for DMD. We previously developed polyethylene glycol (PEG)-modified liposomes (Bubble liposomes) that entrap ultrasound contrast gas and demonstrated that the combination of Bubble liposomes with ultrasound exposure is an effective gene delivery tool in vitro and in vivo. In this study, to evaluate the ability of Bubble liposomes as a PMO delivery tool, we tested the potency of the Bubble liposomes combined with ultrasound exposure to boost the delivery of PMO and increase the skipping of the mutated exon in the mdx mouse. The results indicated that the combination of Bubble liposomes and ultrasound exposure increased the uptake of the PMO targeting a nonsense mutation in exon 23 of the dystrophin gene and consequently increased the PMO-mediated exon-skipping efficiency compared with PMO injection alone, leading to significantly enhanced dystrophin expression. This increased efficiency indicated the potential of the combination of Bubble liposomes with ultrasound exposure to enhance PMO delivery for treating DMD. Thus, this ultrasound-mediated Bubble liposome technique may provide an effective, noninvasive, nonviral method for PMO therapy for DMD muscle as well as for other muscular dystrophies.


Assuntos
Distrofina/antagonistas & inibidores , Técnicas de Transferência de Genes , Morfolinos/administração & dosagem , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso/administração & dosagem , Ultrassom , Animais , Apoptose , Western Blotting , Proliferação de Células , Células Cultivadas , Distrofina/fisiologia , Terapia Genética , Técnicas Imunoenzimáticas , Lipossomos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Morfolinos/farmacologia , Músculo Esquelético/patologia , Músculo Esquelético/efeitos da radiação , Distrofia Muscular de Duchenne/genética , Oligonucleotídeos Antissenso/farmacologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Hum Gene Ther ; 24(5): 479-88, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23521559

RESUMO

Duchenne muscular dystrophy (DMD) is a severe muscle wasting disorder caused by mutations in the DMD gene, affecting 1 in 3500 newborn males. Complete loss of muscle dystrophin protein causes progressive muscle weakness and heart and respiratory failure, leading to premature death. Antisense oligonucleotides (AONs) that bind to complementary sequences of the dystrophin pre-mRNA to induce skipping of the targeted exon by modulating pre-mRNA splicing are promising therapeutic agents for DMD. Such AONs can restore the open reading frame of the DMD gene and produce internally deleted, yet partially functional dystrophin protein isoforms in skeletal muscle. Within the last few years, clinical trials using AONs have made considerable progress demonstrating the restoration of functional dystrophin protein and acceptable safety profiles following both local and systemic delivery in DMD patients. However, improvement of AON delivery and efficacy, along with the development of multiple AONs to treat as many DMD patients as possible needs to be addressed for this approach to fulfill its potential. Here, we review the recent progress made in clinical trials using AONs to treat DMD and discuss the current challenges to the development of AON-based therapy for DMD.


Assuntos
Distrofina/genética , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso/uso terapêutico , Precursores de RNA/genética , Ensaios Clínicos como Assunto , Distrofina/antagonistas & inibidores , Éxons , Terapia Genética/tendências , Humanos , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Oligonucleotídeos Antissenso/genética , Precursores de RNA/antagonistas & inibidores
12.
Dev Biol ; 352(2): 228-42, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21256839

RESUMO

In Drosophila, like in humans, Dystrophin Glycoprotein Complex (DGC) deficiencies cause a life span shortening disease, associated with muscle dysfunction. We performed the first in vivo genetic interaction screen in ageing dystrophic muscles and identified genes that have not been shown before to have a role in the development of muscular dystrophy and interact with dystrophin and/or dystroglycan. Mutations in many of the found interacting genes cause age-dependent morphological and heat-induced physiological defects in muscles, suggesting their importance in the tissue. Majority of them is phylogenetically conserved and implicated in human disorders, mainly tumors and myopathies. Functionally they can be divided into three main categories: proteins involved in communication between muscle and neuron, and interestingly, in mechanical and cellular stress response pathways. Our data show that stress induces muscle degeneration and accelerates age-dependent muscular dystrophy. Dystrophic muscles are already compromised; and as a consequence they are less adaptive and more sensitive to energetic stress and to changes in the ambient temperature. However, only dystroglycan, but not dystrophin deficiency causes extreme myodegeneration induced by energetic stress suggesting that dystroglycan might be a component of the low-energy pathway and act as a transducer of energetic stress in normal and dystrophic muscles.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Drosophila/metabolismo , Distroglicanas/genética , Distroglicanas/metabolismo , Distrofina/genética , Distrofina/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Estresse Fisiológico , Animais , Sequência de Bases , Primers do DNA/genética , Modelos Animais de Doenças , Distroglicanas/antagonistas & inibidores , Distroglicanas/deficiência , Distrofina/antagonistas & inibidores , Distrofina/deficiência , Feminino , Genes de Insetos , Humanos , Masculino , Células Musculares/metabolismo , Distrofia Muscular Animal/etiologia , Mutação , Interferência de RNA , Transdução de Sinais
13.
Brain Res Mol Brain Res ; 118(1-2): 52-9, 2003 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-14559354

RESUMO

Nitric oxide (NO) plays an important role in the pathogenesis of neurodegenerative disease. It has been shown that neuronal NO synthase (nNOS), the enzyme that constitutively produces NO in brain, is a component of the dystrophin-associated protein complex. The absence of dystrophin causes Duchenne muscular dystrophy. Thus, we attempted to study whether or not a decrease of dystrophin expression would induce a modification in nNOS expression in cultured human neurons. Human fetal neuronal cultures were treated with antisense oligonucleotides against different isoforms of dystrophin and the expression of nNOS tested by RT-PCR and immunocytochemistry. Results showed that nNOS mRNA was significantly decreased by about 35% in neurons treated with brain-specific dystrophin (brain Dp427) antisense, whereas iNOS expression was not affected. Accordingly, a decrease in immunostaining for nNOS was observed in antisense treated neurons compared to controls. Expression of neuronal markers, such as bFGF or synaptophysin, was not affected by the same antisense treatment. Astrocytes were not affected by treatment, as shown by utrophin expression, a dystrophin-like protein that was not modified in pure astrocytic cultures. Thus, we conclude that a decrease of dystrophin in human neurons is associated with a decrease of nNOS expression.


Assuntos
Encéfalo/enzimologia , Distrofina/deficiência , Neurônios/enzimologia , Óxido Nítrico Sintase/genética , Óxido Nítrico/metabolismo , Encéfalo/fisiopatologia , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Distrofina/antagonistas & inibidores , Distrofina/metabolismo , Feto , Humanos , Imuno-Histoquímica , Distrofia Muscular de Duchenne/complicações , Distrofia Muscular de Duchenne/enzimologia , Distrofia Muscular de Duchenne/fisiopatologia , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/fisiopatologia , Neurônios/citologia , Óxido Nítrico Sintase/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Isoformas de Proteínas/genética , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo
14.
Tsitologiia ; 44(4): 374-8, 2002.
Artigo em Russo | MEDLINE | ID: mdl-12149782

RESUMO

Human minidystrophin gene (pSG5dys plasmid) and hACR-1 gene (pRc-CMV-10.1 plasmid) were cotransfected by means of "gene-gun" to M. quadriceps femoris of mdx mice. Effects of transfection on dystrophin expression and survival of striated muscle fibres (SMF) were studied on the 21st day after shots. In the control mdx dystrophin-positive muscular fibers [D(+)] SMF and destroyed SMF made 2.1 +/- 0.1 and 2.1 +/- 0.3%, respectively. In mice transfected with pSG5dys plasmid (20 mkg of DNA per mouse), the shares of D(+) SMF and dead SMF raised, respectively, up to 5.6 +/- 1.4 and 4.5 +/- 0.9%. Transfection of mice with pRc-CMV-10.1 (DNA dose is 20 mkg per mouse) reduced the levels of apoptosis in SMF and D(+) SMF level to 1.6 +/- 0.6 and 1.1 +/- 0.4%, respectively. Cotransfection by pSG5dys and pRc-CMV-10.1 plasmids (10 and 10 mkg of each plasmids DNA per mouse) reduced the share of D(+) SMF to 1.1 +/- 0.5% and SMF destruction to 0.9 +/- 0.3%. pSG5dys transfection considerably reduced the share of SMF having peripherally located nuclei, thus indicating a decrease in SMF differentiation level after transfection. Cotransfection of ACR-1 gene and a dystrophin minigene did not suppress further cytodifferentiation of mdx muscle fibers. A conclusion is made that ballistic transfection by hACR-1 gene reduces the level of apoptosis in mdx mice SMF without changing the level of SMF differentiation. The cotransfection of mdx mice muscle by hACR-1 and human minidystrophin gene reduces SMF destruction and supports SMF differentiation, too.


Assuntos
Apoptose , Distrofina/antagonistas & inibidores , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Coxa da Perna , Animais , Apoptose/efeitos dos fármacos , Biolística , Diferenciação Celular/efeitos dos fármacos , Distrofina/biossíntese , Distrofina/genética , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Plasmídeos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...