Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Circ Cardiovasc Genet ; 10(1)2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28196902

RESUMO

BACKGROUND: Hyaluronan (HA) is required for endothelial-to-mesenchymal transition and normal heart development in the mouse. Heart abnormalities in hyaluronidase 2 (HYAL2)-deficient (Hyal2-/- ) mice and humans suggested removal of HA is also important for normal heart development. We have performed longitudinal studies of heart structure and function in Hyal2-/- mice to determine when, and how, HYAL2 deficiency leads to these abnormalities. METHODS AND RESULTS: Echocardiography revealed atrial enlargement, atrial tissue masses, and valvular thickening at 4 weeks of age, as well as diastolic dysfunction that progressed with age, in Hyal2-/- mice. These abnormalities were associated with increased HA, vimentin-positive cells, and fibrosis in Hyal2-/- compared with control mice. Based on the severity of heart dysfunction, acute and chronic groups of Hyal2-/- mice that died at an average of 12 and 25 weeks respectively, were defined. Increased HA levels and mesenchymal cells, but not vascular endothelial growth factor in Hyal2-/- embryonic hearts, suggest that HYAL2 is important to inhibit endothelial-to-mesenchymal transition. Consistent with this, in wild-type embryos, HYAL2 and HA were readily detected, and HA levels decreased with age. CONCLUSIONS: These data demonstrate that disruption of normal HA catabolism in Hyal2-/- mice causes increased HA, which may promote endothelial-to-mesenchymal transition and proliferation of mesenchymal cells. Excess endothelial-to-mesenchymal transition, resulting in increased mesenchymal cells, is the likely cause of morphological heart abnormalities in both humans and mice. In mice, these abnormalities result in progressive and severe diastolic dysfunction, culminating in heart failure.


Assuntos
Cardiopatias Congênitas/enzimologia , Insuficiência Cardíaca/enzimologia , Hialuronoglucosaminidase/deficiência , Células-Tronco Mesenquimais/enzimologia , Disfunção Ventricular Esquerda/enzimologia , Animais , Cardiomegalia/enzimologia , Cardiomegalia/genética , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Proliferação de Células , Progressão da Doença , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Transição Epitelial-Mesenquimal , Fibrose , Proteínas Ligadas por GPI/deficiência , Proteínas Ligadas por GPI/genética , Predisposição Genética para Doença , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Cardiopatias Congênitas/fisiopatologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Doenças das Valvas Cardíacas/enzimologia , Doenças das Valvas Cardíacas/genética , Doenças das Valvas Cardíacas/patologia , Doenças das Valvas Cardíacas/fisiopatologia , Ácido Hialurônico/metabolismo , Hialuronoglucosaminidase/genética , Células-Tronco Mesenquimais/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica , Fenótipo , Volume Sistólico , Fatores de Tempo , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda
2.
Histochem Cell Biol ; 145(1): 53-66, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26515055

RESUMO

Hyaluronidase 2 (HYAL2) is a membrane-anchored protein that is proposed to initiate the degradation of hyaluronan (HA) in the extracellular matrix. The distribution of HYAL2 in tissues, and of HA in tissues lacking HYAL2, is largely unexplored despite the importance of HA metabolism in several disease processes. Herein, we use immunoblot and histochemical analyses to detect HYAL2 and HA in mouse tissues, as well as agarose gel electrophoresis to examine the size of HA. HYAL2 was detected in all tissues that were examined, including the brain. It was localized to the surface and cytoplasm of endothelial cells, as well as specialized epithelial cells in several tissues, including the skin. Accumulated HA, often of higher molecular mass than that in control tissues, was detected in tissues from Hyal2 (-/-) mice. The accumulating HA was located near to where HYAL2 is normally found, although in some tissues, it was distant from the site of HYAL2 localization. Overall, HYAL2 was highest in tissues that remove HA from the circulation (liver, lymph node and spleen), but the levels of HA accumulation in Hyal2 (-/-) mice were highest in tissues that catabolize locally synthesized HA. Our results support HYAL2's role as an extracellular enzyme that initiates HA breakdown in somatic tissues. However, our findings also suggest that HYAL2 contributes to HA degradation through other routes, perhaps as a soluble or secreted form.


Assuntos
Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Ácido Hialurônico/metabolismo , Hialuronoglucosaminidase/biossíntese , Hialuronoglucosaminidase/farmacocinética , Animais , Eletroforese em Gel de Ágar/métodos , Matriz Extracelular/metabolismo , Proteínas Ligadas por GPI/biossíntese , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/farmacocinética , Hialuronoglucosaminidase/genética , Immunoblotting/métodos , Imuno-Histoquímica/métodos , Camundongos , Camundongos Knockout
3.
Mol Ther ; 23(3): 414-22, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25515709

RESUMO

G(M2) gangliosidoses are severe neurodegenerative disorders resulting from a deficiency in ß-hexosaminidase A activity and lacking effective therapies. Using a Sandhoff disease (SD) mouse model (Hexb(-/-)) of the G(M2) gangliosidoses, we tested the potential of systemically delivered adeno-associated virus 9 (AAV9) expressing Hexb cDNA to correct the neurological phenotype. Neonatal or adult SD and normal mice were intravenously injected with AAV9-HexB or -LacZ and monitored for serum ß-hexosaminidase activity, motor function, and survival. Brain G(M2) ganglioside, ß-hexosaminidase activity, and inflammation were assessed at experimental week 43, or an earlier humane end point. SD mice injected with AAV9-LacZ died by 17 weeks of age, whereas all neonatal AAV9-HexB-treated SD mice survived until 43 weeks (P < 0.0001) with only three exhibiting neurological dysfunction. SD mice treated as adults with AAV9-HexB died between 17 and 35 weeks. Neonatal SD-HexB-treated mice had a significant increase in brain ß-hexosaminidase activity, and a reduction in G(M2) ganglioside storage and neuroinflammation compared to adult SD-HexB- and SD-LacZ-treated groups. However, at 43 weeks, 8 of 10 neonatal-HexB injected control and SD mice exhibited liver or lung tumors. This study demonstrates the potential for long-term correction of SD and other G(M2) gangliosidoses through early rAAV9 based systemic gene therapy.


Assuntos
Dependovirus/genética , Gangliosídeo G(M2)/metabolismo , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Doença de Sandhoff/terapia , Cadeia beta da beta-Hexosaminidase/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Encéfalo/enzimologia , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Expressão Gênica , Vetores Genéticos/efeitos adversos , Inflamação/genética , Inflamação/mortalidade , Inflamação/patologia , Inflamação/terapia , Injeções Intravenosas , Óperon Lac , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/patologia , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/patologia , Lisossomos/enzimologia , Lisossomos/patologia , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/genética , Doença de Sandhoff/genética , Doença de Sandhoff/mortalidade , Doença de Sandhoff/patologia , Análise de Sobrevida , Cadeia beta da beta-Hexosaminidase/metabolismo
4.
BBA Clin ; 1: 33-43, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26676230

RESUMO

Bowen-Conradi syndrome (BCS) is a lethal autosomal recessive disorder caused by a D86G substitution in the protein, Essential for Mitotic Growth 1 (EMG1). EMG1 is essential for 18S rRNA maturation and 40S ribosome biogenesis in yeast, but no studies of its role in ribosome biogenesis have been done in mammals. To assess the effect of the EMG1 mutation on cell growth and ribosomal biogenesis in humans, we employed BCS patient cells. The D86G substitution did not interfere with EMG1 nucleolar localization. In BCS patient lymphoblasts, cells accumulated in G2/M, resulting in reduced proliferation rates; however, patient fibroblasts showed normal proliferation. The rate of 18S rRNA processing was consistently delayed in patient cells, although this did not lead to a difference in the levels of 40S ribosomes, or a change in protein synthesis rates. These results demonstrate that as in yeast, EMG1 in mammals has a role in ribosome biogenesis. The obvious phenotype in lymphoblasts compared to fibroblasts suggests a greater need for EMG1 in rapidly dividing cells. Tissue-specific effects have been seen in other ribosomal biogenesis disorders, and it seems likely that the impact of EMG1 deficiency would be larger in the rapidly proliferating cells of the developing embryo.

5.
In. Taller sobre Financiamiento de la Salud en el Proceso de la Reforma del Sector. Taller sobre Financiamiento de la Salud en el Proceso de la Reforma del Sector / Workshop on Health Care Financing in the Process of Health Sector Reform. Washington, D.C, Organización Panamericana de la Salud. Programa de Políticas Públicas y Salud, 1995. p.19-25, tab.
Monografia em Inglês | LILACS | ID: lil-375214
6.
In. Taller sobre Financiamiento de la Salud en el Proceso de la Reforma del Sector. Taller sobre Financiamiento de la Salud en el Proceso de la Reforma del Sector / Workshop on Health Care Financing in the Process of Health Sector Reform. Washington, D.C, Organización Panamericana de la Salud. Programa de Políticas Públicas y Salud, 1995. p.119-128, ilus.
Monografia em Inglês | LILACS | ID: lil-375226
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA