Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 9674, 2022 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-35690627

RESUMO

Duchenne muscular dystrophy is a severe neuromuscular disease causing a progressive muscle wasting due to mutations in the DMD gene that lead to the absence of dystrophin protein. Adeno-associated virus (AAV)-based therapies aiming to restore dystrophin in muscles, by either exon skipping or microdystrophin expression, are very promising. However, the absence of dystrophin induces cellular perturbations that hinder AAV therapy efficiency. We focused here on the impact of the necrosis-regeneration process leading to nuclear centralization in myofiber, a common feature of human myopathies, on AAV transduction efficiency. We generated centronucleated myofibers by cardiotoxin injection in wild-type muscles prior to AAV injection. Intramuscular injections of AAV1 vectors show that transgene expression was drastically reduced in regenerated muscles, even when the AAV injection occurred 10 months post-regeneration. We show also that AAV genomes were not lost from cardiotoxin regenerated muscle and were properly localised in the myofiber nuclei but were less transcribed leading to muscle transduction defect. A similar defect was observed in muscles of the DMD mouse model mdx. Therefore, the regeneration process per se could participate to the AAV-mediated transduction defect observed in dystrophic muscles which may limit AAV-based therapies.


Assuntos
Distrofia Muscular Animal , Distrofia Muscular de Duchenne , Animais , Cardiotoxinas/farmacologia , Dependovirus/genética , Dependovirus/metabolismo , Distrofina/genética , Distrofina/metabolismo , Terapia Genética , Vetores Genéticos/genética , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Regeneração/genética , Transgenes
2.
EMBO Mol Med ; 14(5): e12860, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35298089

RESUMO

Duchenne muscular dystrophy (DMD) is characterized by progressive muscle degeneration. Two important deleterious features are a Ca2+ dysregulation linked to Ca2+ influxes associated with ryanodine receptor hyperactivation, and a muscular nicotinamide adenine dinucleotide (NAD+ ) deficit. Here, we identified that deletion in mdx mice of CD38, a NAD+ glycohydrolase-producing modulators of Ca2+ signaling, led to a fully restored heart function and structure, with skeletal muscle performance improvements, associated with a reduction in inflammation and senescence markers. Muscle NAD+ levels were also fully restored, while the levels of the two main products of CD38, nicotinamide and ADP-ribose, were reduced, in heart, diaphragm, and limb. In cardiomyocytes from mdx/CD38-/- mice, the pathological spontaneous Ca2+ activity was reduced, as well as in myotubes from DMD patients treated with isatuximab (SARCLISA® ) a monoclonal anti-CD38 antibody. Finally, treatment of mdx and utrophin-dystrophin-deficient (mdx/utr-/- ) mice with CD38 inhibitors resulted in improved skeletal muscle performances. Thus, we demonstrate that CD38 actively contributes to DMD physiopathology. We propose that a selective anti-CD38 therapeutic intervention could be highly relevant to develop for DMD patients.


Assuntos
Distrofia Muscular de Duchenne , ADP-Ribosil Ciclase 1 , Animais , Humanos , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético , Distrofia Muscular de Duchenne/genética , Miócitos Cardíacos/patologia , NAD/genética , NAD/uso terapêutico , NAD+ Nucleosidase/genética , Fenótipo
3.
Mol Ther Methods Clin Dev ; 17: 695-708, 2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32346547

RESUMO

Duchenne muscular dystrophy (DMD) is a devastating neuromuscular disease caused by an absence of the dystrophin protein, which is essential for muscle fiber integrity. Among the developed therapeutic strategies for DMD, the exon-skipping approach corrects the frameshift and partially restores dystrophin expression. It could be achieved through the use of antisense sequences, such as peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO) or the small nuclear RNA-U7 carried by an adeno-associated virus (AAV) vector. AAV-based gene therapy approaches have potential for use in DMD treatment but are subject to a major limitation: loss of the AAV genome, necessitating readministration of the vector, which is not currently possible, due to the immunogenicity of the capsid. The PPMO approach requires repeated administrations and results in only weak cardiac dystrophin expression. Here, we evaluated a combination of PPMO- and AAV-based therapy in a mouse model of severe DMD. Striking benefits of this combined therapy were observed in striated muscles, with marked improvements in heart and diaphragm structure and function, with unrivalled extent of survival, opening novel therapeutic perspectives for patients.

4.
Sci Rep ; 9(1): 1808, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755642

RESUMO

PiT1/SLC20A1 is an inorganic phosphate transporter with additional functions including the regulation of TNFα-induced apoptosis, erythropoiesis, cell proliferation and insulin signaling. Recent data suggest a relationship between PiT1 and NF-κB-dependent inflammation: (i) Pit1 mRNA is up-regulated in the context of NF-κB pathway activation; (ii) NF-κB target gene transcription is decreased in PiT1-deficient conditions. This led us to investigate the role of PiT1 in lipopolysaccharide (LPS)-induced inflammation. MCP-1 and IL-6 concentrations were impaired in PiT1-deficient bone marrow derived macrophages (BMDMs) upon LPS stimulation. Lower MCP-1 and IL-6 serum levels were observed in Mx1-Cre; Pit1lox/lox mice dosed intraperitoneally with LPS. Lower PiT1 expression correlated with decreased in vitro wound healing and lower reactive oxygen species levels. Reduced IκB degradation and lower p65 nuclear translocation were observed in PiT1-deficient cells stimulated with LPS. Conversely, PiT1 expression was induced in vitro upon LPS stimulation. Addition of an NF-κB inhibitor abolished LPS-induced PiT1 expression. Furthermore, we showed that p65 expression activated Pit1 promoter activity. Finally, ChIP assays demonstrated that p65 directly binds to the mPit1 promoter in response to LPS. These data demonstrate a completely novel function of PiT1 in the response to LPS and provide mechanistic insights into the regulation of PiT1 expression by NF-κB.


Assuntos
Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Fator de Transcrição Pit-1/metabolismo , Animais , Apoptose/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , NADPH Oxidase 2/metabolismo , NF-kappa B/metabolismo , Peritonite/induzido quimicamente , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tioglicolatos/toxicidade , Fator de Transcrição Pit-1/genética , Fator de Necrose Tumoral alfa/metabolismo , Cicatrização/efeitos dos fármacos
5.
Cell Rep ; 16(10): 2736-2748, 2016 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-27568561

RESUMO

The liver plays a central role in whole-body lipid and glucose homeostasis. Increasing dietary fat intake results in increased hepatic fat deposition, which is associated with a risk for development of insulin resistance and type 2 diabetes. In this study, we demonstrate a role for the phosphate inorganic transporter 1 (PiT1/SLC20A1) in regulating metabolism. Specific knockout of Pit1 in hepatocytes significantly improved glucose tolerance and insulin sensitivity, enhanced insulin signaling, and decreased hepatic lipogenesis. We identified USP7 as a PiT1 binding partner and demonstrated that Pit1 deletion inhibited USP7/IRS1 dissociation upon insulin stimulation. This prevented IRS1 ubiquitination and its subsequent proteasomal degradation. As a consequence, delayed insulin negative feedback loop and sustained insulin signaling were observed. Moreover, PiT1-deficient mice were protected against high-fat-diet-induced obesity and diabetes. Our findings indicate that PiT1 has potential as a therapeutic target in the context of metabolic syndrome, obesity, and diabetes.


Assuntos
Glucose/metabolismo , Hepatócitos/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Insulina/metabolismo , Transdução de Sinais , Fator de Transcrição Pit-1/metabolismo , Peptidase 7 Específica de Ubiquitina/metabolismo , Tecido Adiposo/patologia , Envelhecimento/patologia , Animais , Dieta Hiperlipídica , Fígado Gorduroso/complicações , Fígado Gorduroso/patologia , Fibroblastos/metabolismo , Gluconeogênese , Teste de Tolerância a Glucose , Inflamação/complicações , Inflamação/patologia , Resistência à Insulina , Camundongos Knockout , Obesidade/patologia , Especificidade de Órgãos , Fenótipo , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Ubiquitinação , Aumento de Peso
6.
Blood ; 121(4): 666-78, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23190530

RESUMO

The PIT1/SLC20A1 protein, a well-described sodium/phosphate cotransporter and retrovirus receptor, has been identified recently as a modular of proliferation and apoptosis in vitro. The targeted deletion of the PIT1 gene in mice revealed a lethal phenotype due to severe anemia attributed to defects in liver development. However, the presence of immature erythroid cells associated with impaired maturation of the globin switch led us to investigate the role of PIT1 in hematopoietic development. In the present study, specific deletion of PIT1 in the hematopoietic system and fetal liver transplantation experiments demonstrated that anemia was associated with an erythroid cell- autonomous defect. Moreover, anemia was not due to RBC destruction but rather to maturation defects. Because Erythroid Krüppel-like Factor (EKLF)-knockout mice showed similar maturation defects, we investigated the functional link between PIT1 and EKLF. We demonstrated that EKLF increases PIT1 expression during RBC maturation by binding to its promoter in vivo and that shRNA-driven depletion of either PIT1 or EKLF impairs erythroid maturation of G1E cells in vitro, whereas reexpression of PIT1 in EKLF-depleted G1E cells partially restores erythroid maturation. This is the first demonstration of a physiologic involvement of PIT1 in erythroid maturation in vivo.


Assuntos
Células Eritroides/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fator de Transcrição Pit-1/genética , Animais , Sequência de Bases , Diferenciação Celular , Ensaio de Unidades Formadoras de Colônias , Células Eritroides/citologia , Eritropoese/genética , Deleção de Genes , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Fígado/embriologia , Fígado/metabolismo , Camundongos , Dados de Sequência Molecular , Fenótipo , Regiões Promotoras Genéticas , Alinhamento de Sequência , Ativação Transcricional
7.
PLoS One ; 5(2): e9148, 2010 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-20161774

RESUMO

BACKGROUND: PiT1 (or SLC20a1) encodes a widely expressed plasma membrane protein functioning as a high-affinity Na(+)-phosphate (Pi) cotransporter. As such, PiT1 is often considered as a ubiquitous supplier of Pi for cellular needs regardless of the lack of experimental data. Although the importance of PiT1 in mineralizing processes have been demonstrated in vitro in osteoblasts, chondrocytes and vascular smooth muscle cells, in vivo evidence is missing. METHODOLOGY/PRINCIPAL FINDINGS: To determine the in vivo function of PiT1, we generated an allelic series of PiT1 mutations in mice by combination of wild-type, hypomorphic and null PiT1 alleles expressing from 100% to 0% of PiT1. In this report we show that complete deletion of PiT1 results in embryonic lethality at E12.5. PiT1-deficient embryos display severely hypoplastic fetal livers and subsequent reduced hematopoiesis resulting in embryonic death from anemia. We show that the anemia is not due to placental, yolk sac or vascular defects and that hematopoietic progenitors have no cell-autonomous defects in proliferation and differentiation. In contrast, mutant fetal livers display decreased proliferation and massive apoptosis. Animals carrying two copies of hypomorphic PiT1 alleles (resulting in 15% PiT1 expression comparing to wild-type animals) survive at birth but are growth-retarded and anemic. The combination of both hypomorphic and null alleles in heterozygous compounds results in late embryonic lethality (E14.5-E16.5) with phenotypic features intermediate between null and hypomorphic mice. In the three mouse lines generated we could not evidence defects in early skeleton formation. CONCLUSION/SIGNIFICANCE: This work is the first to illustrate a specific in vivo role for PiT1 by uncovering it as being a critical gene for normal developmental liver growth.


Assuntos
Embrião de Mamíferos/metabolismo , Fígado/metabolismo , Mutação , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Animais , Apoptose , Contagem de Células , Proliferação de Células , Células Cultivadas , Embrião de Mamíferos/anormalidades , Eritrócitos/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes Essenciais , Genótipo , Células-Tronco Hematopoéticas/metabolismo , Fígado/citologia , Fígado/embriologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/deficiência , Fatores de Tempo
8.
Cell Res ; 19(8): 1018-30, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19417777

RESUMO

Neonatal gonocytes are precursors of spermatogonial stem cells. Preserving their integrity by elimination of damaged germ cells may be crucial to avoid the transmission of genetic alterations to progeny. Using gamma-irradiation, we investigated by immunohistochemistry, flow cytometry and real-time PCR components of the death machinery in neonatal gonocytes. Their death was correlated with caspase 3 activation but not with AIF translocation into the nucleus. The in vivo contribution of both the extrinsic and the intrinsic pathways was then investigated. We focused on the roles of TRAIL/Death Receptor 5 (DR5) and PUMA. Our results were validated using knockout mice. Whereas DR5 expression was upregulated at the cell surface after radiation, caspase 8 was not activated. However, we detected caspase 9 cleavage associated with cytochrome c release. In mice deficient for PUMA, radiation-induced gonocyte apoptosis was reduced, whereas invalidation of TRAIL had no effect. Overall, our results show that genotoxic stress-induced apoptosis of gonocytes is caspase-dependent and involves almost exclusively the intrinsic pathway. Furthermore, PUMA plays a critical role in the maintenance of genomic integrity of spermatogonial stem cell precursors.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Espermatogônias/citologia , Células-Tronco/citologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Apoptose , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Caspase 9/metabolismo , Citocromos c/metabolismo , Raios gama , Instabilidade Genômica , Masculino , Camundongos , Camundongos Knockout , Fosforilação , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/deficiência , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Testículo/embriologia , Testículo/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
9.
Biol Reprod ; 71(2): 643-9, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15115728

RESUMO

The DNA double-strand breaks (DSBs) are considered to be the most relevant lesions for the deleterious effects of ionizing radiation exposure. The discovery that the induction of DSBs is rapidly followed by the phosphorylation of H2AX histone at Ser-139, favoring repair protein recruitment or access, opens the possibility for a wide range of research. This phosphorylated histone, named gamma-H2AX, has been shown to form foci in interphase nuclei as well as megabase chromatin domains surrounding the DNA lesion on chromosomes. Using detection of gamma-H2AX on germ cell mitotic chromosomes 2 h after gamma-irradiation, we studied radiation-induced DSBs during the G(2)/M phase of the cell cycle. We show that 1) non-irradiated neonatal germ cells express gamma-H2AX with variable patterns at metaphase, 2) gamma-irradiation induces foci whose number increases in a dose-dependent manner, 3) some foci correspond to visible chromatid breaks or exchanges, 4) sticky chromosomes characterizing cell radiation exposure during mitosis are a consequence of DSBs, and 5) gamma-H2AX remains localized at the sites of the lesions even after end-joining has taken place. This suggests that completion of DSB repair does not necessarily imply disappearance of gamma-H2AX.


Assuntos
Cromátides/efeitos da radiação , Aberrações Cromossômicas , DNA/efeitos da radiação , Histonas/genética , Espermatogônias/efeitos da radiação , Animais , Animais Recém-Nascidos , Corantes Azur , Relação Dose-Resposta à Radiação , Raios gama , Expressão Gênica/efeitos da radiação , Masculino , Metáfase , Camundongos , Camundongos Endogâmicos , Troca de Cromátide Irmã/efeitos da radiação , Espermatogênese/genética , Espermatogênese/efeitos da radiação , Espermatogônias/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA