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










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 181(9): 6337-48, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18941225

RESUMO

The details of the mechanism by which severe acute respiratory syndrome-associated coronavirus (SARS-CoV) causes severe pneumonia are unclear. We investigated the immune responses and pathologies of SARS-CoV-infected BALB/c mice that were immunized intradermally with recombinant vaccinia virus (VV) that expressed either the SARS-CoV spike (S) protein (LC16m8rVV-S) or simultaneously all the structural proteins, including the nucleocapsid (N), membrane (M), envelope (E), and S proteins (LC16m8rVV-NMES) 7-8 wk before intranasal SARS-CoV infection. The LC16m8rVV-NMES-immunized group exhibited as severe pneumonia as the control groups, although LC16m8rVV-NMES significantly decreased the pulmonary SARS-CoV titer to the same extent as LC16m8rVV-S. To identify the cause of the exacerbated pneumonia, BALB/c mice were immunized with recombinant VV that expressed the individual structural proteins of SARS-CoV (LC16mOrVV-N, -M, -E, -S) with or without LC16mOrVV-S (i.e., LC16mOrVV-N, LC16mOrVV-M, LC16mOrVV-E, or LC16mOrVV-S alone or LC16mOrVV-N + LC16mOrVV-S, LC16mOrVV-M + LC16mOrVV-S, or LC16mOrVV-E + LC16mOrVV-S), and infected with SARS-CoV more than 4 wk later. Both LC16mOrVV-N-immunized mice and LC16mOrVV-N + LC16mOrVV-S-immunized mice exhibited severe pneumonia. Furthermore, LC16mOrVV-N-immunized mice upon infection exhibited significant up-regulation of both Th1 (IFN-gamma, IL-2) and Th2 (IL-4, IL-5) cytokines and down-regulation of anti-inflammatory cytokines (IL-10, TGF-beta), resulting in robust infiltration of neutrophils, eosinophils, and lymphocytes into the lung, as well as thickening of the alveolar epithelium. These results suggest that an excessive host immune response against the nucleocapsid protein of SARS-CoV is involved in severe pneumonia caused by SARS-CoV infection. These findings increase our understanding of the pathogenesis of SARS.


Assuntos
Proteínas do Nucleocapsídeo/imunologia , Pneumonia Viral/imunologia , Síndrome Respiratória Aguda Grave/prevenção & controle , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/biossíntese , Linhagem Celular , Chlorocebus aethiops , Proteínas do Nucleocapsídeo de Coronavírus , Citocinas/biossíntese , Feminino , Humanos , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Nucleocapsídeo/administração & dosagem , Pneumonia Viral/patologia , Pneumonia Viral/virologia , RNA Mensageiro/biossíntese , Coelhos , Síndrome Respiratória Aguda Grave/patologia , Síndrome Respiratória Aguda Grave/virologia , Índice de Gravidade de Doença , Células Vero , Vacinas Virais/administração & dosagem
2.
Clin Chim Acta ; 360(1-2): 103-7, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16112661

RESUMO

BACKGROUND: Fabry disease is an X-linked lysosomal storage disease resulting from deficient activity of the enzyme alpha-galacotsidase A. Accumulation of glycosphingolipids, especially globotriaosylceramide, leads to renal damage in Fabry disease. In patients with Fabry disease, the urinary sediment contains excreted glycosphingolipids. With enzyme replacement therapy for Fabry disease now currently available, we examined whether the urinary sediment could be used to noninvasively monitor effectiveness of enzyme replacement therapy. METHODS: Four male patients with hemizygous classical Fabry disease received recombinant alpha-galactsidase A biweekly, and urinary sediments were assessed at 3-month intervals. RESULTS: The morphologic and immunohistochemical changes in urinary sediment at 6 and 18 months suggested that accumulations of glycosphingolipids in renal tissues were cleared by enzyme replacement. CONCLUSION: Examination of urinary sediments could serve as noninvasive monitoring of the effect of therapy in patients with Fabry disease.


Assuntos
Monitoramento de Medicamentos/métodos , Doença de Fabry/tratamento farmacológico , Doença de Fabry/urina , alfa-Galactosidase/administração & dosagem , Adulto , Terapia Enzimática , Glicoesfingolipídeos/urina , Humanos , Imuno-Histoquímica , Masculino , Urina/química , Urina/citologia
3.
Pediatr Int ; 46(6): 673-7, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15660866

RESUMO

BACKGROUND: Human alpha-galactosidase A (halphaG) is an essential lysosomal enzyme in catalyzing the hydrolysis of ceramide trihexoside in humans. Effects have been directed to develop effective gene and replacement therapies for the deficiency of halphaG, Fabry disease. In recent years, halphaG transgenic mice (TGM) have been established, and the expression of halphaG in their general organs has been reported. However, detailed distribution of the cells expressing halphaG have not yet been defined. METHODS: The distribution of halphaG in organs of the halphaG-TGM was studied by means of immunohistochemistry and enzyme assay. RESULTS: Immunohistochemical analysis revealed a systematic halphaG expression in the TGM, including endothelial cells of the bone marrow, liver, spleen, pancreas, lungs, uriniferous tubules in the kidneys, and choroids plexus in the brain. Enzyme assay demonstrated a persistent expression of halphaG in the TGM during 14-20 months after birth. CONCLUSION: A long-term expression of halphaG in organs may indicate halphaG-TGM as a useful tool in the research of gene and replacement therapies for Fabry disease.


Assuntos
Doença de Fabry/enzimologia , alfa-Galactosidase/metabolismo , Animais , Biópsia por Agulha , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Doença de Fabry/genética , Doença de Fabry/terapia , Feminino , Previsões , Regulação da Expressão Gênica , Terapia Genética/normas , Terapia Genética/tendências , Vetores Genéticos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/genética , Sensibilidade e Especificidade , alfa-Galactosidase/genética
4.
Mol Genet Metab ; 78(1): 51-8, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12559848

RESUMO

G(M1)-gangliosidosis and Morquio B disease are distinct in clinical and biochemical features, but both disorders are caused by genetic defects of the same enzyme, acid beta-galactosidase (beta-Gal). We analyzed the kinetic properties of mutant beta-Gals from patients with G(M1)-gangliosidosis and Morquio B disease to examine the clinical and biochemical differences between both disorders. Five skin fibroblast lines from patients with G(M1)-gangliosidosis (2 cases; R201C/R201C and I51T/I51T), Morquio B disease (2 cases; W273L/W273L and Y83H/R482C), and galactosialidosis (1 case; Y395C/S90L) were used as enzyme sources. Residual enzyme activity in the cells was subjected to kinetic analysis. Substrate analogs including Galbeta1-3GalNAc, as an analog for G(M1)-ganglioside, and Galbeta1-4GlcNAc, as an analog for keratan sulfate, were used to determine IC(50) and K(i) for beta-Gals with an artificial substrate (4-methylumbelliferyl beta-D-galactopyranoside). Enzymatic assay method was established to examine the hydrolytic activity with the mutant beta-Gal for the substrate analogs. The mutant beta-Gal activities were inhibited by Galbeta1-3GalNAc and Galbeta1-4GlcNAc in a concentration-dependent manner. Remarkable increase in IC(50) ratio and K(i) ratio (Galbeta1-4GlcNAc/Galbeta1-3GalNAc) was observed in Morquio B disease. Relative hydrolytic activity (Galbeta1-4GlcNAc/Galbeta1-3GalNAc) was markedly decreased in Morquio B disease as compared with other subjects; controls (means+/-SD, n=4), 1.00+/-0.02; galactosialidosis, 1.03; G(M1)-gangliosidosis, 1.15 and 1.00; and Morquio B disease, 0.27 and 0.32. The mutant beta-Gals from the patients with Morquio B disease exhibited lower affinity and lower hydrolytic activity toward Galbeta1-4GlcNAc rather than Galbeta1-3GalNAc. These findings suggest that imbalanced substrate specificity of the mutant beta-Gals induces predominant accumulation of keratan sulfate and a rationale for performing differential diagnostic analysis for both disorders.


Assuntos
Mucopolissacaridose IV/enzimologia , beta-Galactosidase/metabolismo , Antígenos Glicosídicos Associados a Tumores/metabolismo , Antígenos Glicosídicos Associados a Tumores/farmacologia , Linhagem Celular , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/farmacologia , Humanos , Hidrólise , Sulfato de Queratano/análogos & derivados , Sulfato de Queratano/metabolismo , Sulfato de Queratano/farmacologia , Cinética , Mucopolissacaridose IV/genética , Mucopolissacaridose IV/patologia , Mutação , Especificidade por Substrato , beta-Galactosidase/antagonistas & inibidores , beta-Galactosidase/genética
5.
Glycobiology ; 12(12): 821-8, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12499404

RESUMO

A mammalian-like sugar moiety was created in glycoprotein by Saccharomyces cerevisiae in combination with bacterial alpha-mannosidase to produce a more economic enzyme replacement therapy for patients with Fabry disease. We introduced the human alpha-galactosidase A (alpha-GalA) gene into an S. cerevisiae mutant that was deficient in the outer chains of N-linked mannan. The recombinant alpha-GalA contained both neutral (Man(8)GlcNAc(2)) and acidic ([Man-P](1-2)Man(8)GlcNAc(2)) sugar chains. Because an efficient incorporation of alpha-GalA into lysosomes of human cells requires mannose-6-phosphate (Man-6-P) residues that should be recognized by the specific receptor, we trimmed down the sugar chains of the alpha-GalA by a newly isolated bacterial alpha-mannosidase. Treatment of the alpha-GalA with the alpha-mannosidase resulted in the exposure of a Man-6-P residue on a nonreduced end of oligosaccharide chains after the removal of phosphodiester-linked nonreduced-end mannose. The treated alpha-GalA was efficiently incorporated into fibroblasts derived from patients with Fabry disease. The uptake was three to four times higher than that of the nontreated alpha-GalA and was inhibited by the addition of 5 mM Man-6-P. Incorporated alpha-GalA was targeted to the lysosome, and hydrolyzed ceramide trihexoside accumulated in the Fabry fibroblasts after 5 days. This method provides an effective and economic therapy for many lysosomal disorders, including Fabry disease.


Assuntos
Doença de Fabry/tratamento farmacológico , Lisossomos/enzimologia , alfa-Galactosidase/genética , Sequência de Carboidratos , Clonagem Molecular , Humanos , Dados de Sequência Molecular , Oligossacarídeos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapêutico , Saccharomyces cerevisiae/enzimologia , alfa-Galactosidase/química , alfa-Galactosidase/uso terapêutico
6.
Proc Natl Acad Sci U S A ; 99(21): 13777-82, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12370426

RESUMO

Fabry disease is a systemic disease caused by genetic deficiency of a lysosomal enzyme, alpha-galactosidase A (alpha-gal A), and is thought to be an important target for enzyme replacement therapy. We studied the feasibility of gene-mediated enzyme replacement for Fabry disease. The adeno-associated virus (AAV) vector containing the alpha-gal A gene was injected into the right quadriceps muscles of Fabry knockout mice. A time course study showed that alpha-gal A activity in plasma was increased to approximately 25% of normal mice and that this elevated activity persisted for up to at least 30 weeks without development of anti-alpha-gal A antibodies. The alpha-gal A activity in various organs of treated Fabry mice remained 5-20% of those observed in normal mice. Accumulated globotriaosylceramide in these organs was completely cleared by 25 weeks after vector injection. Reduction of globotriaosylceramide levels was also confirmed by immunohistochemical and electronmicroscopic analyses. Echocardiographic examination of treated mice demonstrated structural improvement of cardiac hypertrophy 25 weeks after the treatment. AAV vector-mediated muscle-directed gene transfer provides an efficient and practical therapeutic approach for Fabry disease.


Assuntos
Doença de Fabry/genética , Doença de Fabry/terapia , Terapia Genética/métodos , Animais , Linhagem Celular , Dependovirus/genética , Modelos Animais de Doenças , Doença de Fabry/enzimologia , Doença de Fabry/patologia , Feminino , Vetores Genéticos , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/enzimologia , Miocárdio/patologia , Triexosilceramidas/metabolismo , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...