Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 317
Filtrar
1.
Clin Genet ; 71(3): 260-6, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17309649

RESUMO

Interstitial deletions of the long arm of chromosome 6 are relatively rare, with fewer than 100 cases reported. Phenotypic variation is in large part due to differences in size and location of the segmental aneuploidy. We report three new patients with interstitial deletions of chromosome 6q defined at the molecular level by array comparative genomic hybridization (array CGH). In two of three cases, the molecular breakpoints differed from those indicated by conventional karyotyping, demonstrating the enhanced resolution of array CGH. Two patients had minimal deletions of 6 and 8.8 Mb involving 6q16.2-->q21, and the third patient had a deletion of 11.3 Mb spanning 6q15-->q21. All three had developmental delay, craniofacial dysmorphology, and functional eye disorders, suggesting that genes affecting brain and craniofacial development are located in 6q16.2-->q21, the deleted region common to all three patients. Furthermore, gene(s) for discordant phenotypic features, such as central diabetes insipidus, may reside at 6q15, the monosomic region unique to patient 3. All three cases described here showed loss of paternal alleles within the deleted segment, providing further evidence of the predominantly paternal origin for 6q deletions and rearrangements.


Assuntos
Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Deleção Cromossômica , Cromossomos Humanos Par 6 , Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/genética , Adolescente , Aneuploidia , Criança , Análise Citogenética , Feminino , Genótipo , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Fenótipo
2.
Hum Mol Genet ; 14(3): 373-84, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15590701

RESUMO

The central nervous system of persons with Down syndrome presents cytoarchitectural abnormalities that likely result from gene-dosage effects affecting the expression of key developmental genes. To test this hypothesis, we have investigated the transcriptome of the cerebellum of the Ts1Cje mouse model of Down syndrome during postnatal development using microarrays and quantitative PCR (qPCR). Genes present in three copies were consistently overexpressed, with a mean ratio relative to euploid of 1.52 as determined by qPCR. Out of 63 three-copy genes tested, only five, nine and seven genes had ratios >2 or <1.2 at postnatal days 0 (P0), P15 and P30, respectively. This gene-dosage effect was associated with a dysregulation of the expression of some two-copy genes. Out of 8258 genes examined, the Ts1Cje/euploid ratios differed significantly from 1.0 for 406 (80 and 154 with ratios above 1.5 and below 0.7, respectively), 333 (11 above 1.5 and 55 below 0.7) and 246 genes (59 above 1.5 and 69 below 0.7) at P0, P15 and P30, respectively. Among the two-copy genes differentially expressed in the trisomic cerebellum, six homeobox genes, two belonging to the Notch pathway, were severely repressed. Overall, at P0, transcripts involved in cell differentiation and development were over-represented among the dysregulated genes, suggesting that cell differentiation and migration might be more altered than cell proliferation. Finally, global gene profiling revealed that transcription in Ts1Cje mice is more affected by the developmental changes than by the trisomic state, and that there is no apparent detectable delay in the postnatal development of the cerebellum of Ts1Cje mice.


Assuntos
Cerebelo/metabolismo , Síndrome de Down/genética , Perfilação da Expressão Gênica , Animais , Diferenciação Celular , Cerebelo/crescimento & desenvolvimento , Modelos Animais de Doenças , Síndrome de Down/metabolismo , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Análise de Componente Principal
3.
Dev Dyn ; 232(1): 131-9, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15580624

RESUMO

To investigate the genetic contribution to phenotypic variability in aneuploidy, we generated mice with trisomy 16 (Ts16) by mating [Rb(6.16)24Lub x Rb(16.17)7Bnr]F1 males with females from four inbred strains, BALB/cJ, C3H/HeJ, C57BL/6J, and DBA/2J. Among the four Ts16 strains that were generated, there were no significant differences in survival, weight, or length relative to euploid control littermates at either embryonic day (E) 14.5 or E17.5. All Ts16 fetuses at E14.5 had edema that ranged from mild to severe, increased amniotic fluid volume, and a thickened neck. At E17.5, Ts16 fetuses exhibited two distinct phenotypes, one with an edematous morphology and the other runt-like. None of these gross morphological abnormalities was strain-specific either in occurrence or frequency. At E10.5, there were pharyngeal arch artery (PAA) anomalies in all Ts16 embryos on the C3H/HeJ background, but none in trisomics on the other three backgrounds. However, at E17.5, there was in addition to ventricular and atrioventricular septal defects, a high frequency of aortic arch defects in Ts16 fetuses, irrespective of genetic background. Taken together, these findings indicate that there are at least two mechanistic responses to the presence of three copies of mouse chromosome 16 in the modeling of the cardiovascular system: one, development of PAA defects, is strongly influenced by genetic background; but the second, development of aortic arch anomalies in the absence of preexisting PAA anomalies, is not.


Assuntos
Camundongos Mutantes , Camundongos Transgênicos , Trissomia , Animais , Artérias/anormalidades , Região Branquial/irrigação sanguínea , Sistema Cardiovascular/embriologia , Cruzamentos Genéticos , Edema/genética , Feminino , Coração/embriologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Fenótipo , Especificidade da Espécie , Fatores de Tempo
4.
Dev Dyn ; 230(3): 581-9, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15188443

RESUMO

Two mouse models are widely used for Down syndrome (DS) research. The Ts65Dn mouse carries a small chromosome derived primarily from mouse chromosome 16, causing dosage imbalance for approximately half of human chromosome 21 orthologs. These mice have cerebellar pathology with direct parallels to DS. The Ts1Cje mouse, containing a translocated chromosome 16, is at dosage imbalance for 67% of the genes triplicated in Ts65Dn. We quantified cerebellar volume and granule cell and Purkinje cell density in Ts1Cje. Cerebellar volume was significantly affected to the same degree in Ts1Cje and Ts65Dn, despite that Ts1Cje has fewer triplicated genes. However, dosage imbalance in Ts1Cje had little effect on granule cell and Purkinje cell density. Several mice with dosage imbalance for the segment of the Ts65Dn chromosome not triplicated in Ts1Cje had phenotypes that contrasted with those in Ts1Cje. These observations do not readily differentiate between two prevalent hypotheses for gene action in DS.


Assuntos
Cerebelo/patologia , Modelos Animais de Doenças , Síndrome de Down/genética , Síndrome de Down/patologia , Fenótipo , Animais , Cerebelo/diagnóstico por imagem , Cruzamentos Genéticos , Feminino , Marcadores Genéticos , Granulócitos/patologia , Humanos , Imageamento Tridimensional , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Biossíntese de Proteínas , Células de Purkinje/patologia , Deleção de Sequência , Trissomia , Ultrassonografia
5.
Am J Physiol Cell Physiol ; 286(5): C1152-8, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15075214

RESUMO

Contractions of skeletal muscles produce increases in concentrations of superoxide anions and activity of hydroxyl radicals in the extracellular space. The sources of these reactive oxygen species are not clear. We tested the hypothesis that, after a demanding isometric contraction protocol, the major source of superoxide and hydroxyl radical activity in the extracellular space of muscles is mitochondrial generation of superoxide anions and that, with a reduction in MnSOD activity, concentration of superoxide anions in the extracellular space is unchanged but concentration of hydroxyl radicals is decreased. For gastrocnemius muscles from adult (6-8 mo old) wild-type (Sod2(+/+)) mice and knockout mice heterozygous for the MnSOD gene (Sod2(+/-)), concentrations of superoxide anions and hydroxyl radical activity were measured in the extracellular space by microdialysis. A 15-min protocol of 180 isometric contractions induced a rapid, equivalent increase in reduction of cytochrome c as an index of superoxide anion concentrations in the extracellular space of Sod2(+/+) and Sod2(+/-) mice, whereas hydroxyl radical activity measured by formation of 2,3-dihydroxybenzoate from salicylate increased only in the extracellular space of muscles of Sod2(+/+) mice. The lack of a difference in increase in superoxide anion concentration in the extracellular space of Sod2(+/+) and Sod2(+/-) mice after the contraction protocol supported the hypothesis that superoxide anions were not directly derived from mitochondria. In contrast, the data obtained suggest that the increase in hydroxyl radical concentration in the extracellular space of muscles from wild-type mice after the contraction protocol most likely results from degradation of hydrogen peroxide generated by MnSOD activity.


Assuntos
Espaço Extracelular/metabolismo , Contração Isométrica/fisiologia , Mitocôndrias Musculares/enzimologia , Músculo Esquelético/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/fisiologia , Animais , Citocromos c/metabolismo , Heterozigoto , Hidroxibenzoatos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Músculo Esquelético/fisiologia , Superóxido Dismutase/genética
6.
Free Radic Biol Med ; 31(9): 1101-10, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11677043

RESUMO

Mn superoxide dismutase (MnSOD), a mitochondrial antioxidant enzyme, has been shown to be essential for animal survival. MnSOD mutant mice (Sod2-/- mice) on the CD1 background develop severe dilated cardiomyopathy and usually die within 10 d after birth. To characterize better the phenotype and understand the mechanism of superoxide-mediated tissue damage in Sod2-/- mice, congenic Sod2-/- mice on inbred backgrounds were generated to ensure genetic homogeneity. When generated on a C57BL/6J background (B6), more than half of the fetuses develop severe dilated cardiomyopathy by embryonic day 15 and die in the uterus. Those that survive to term usually die within 24 h. In contrast, Sod2-/- mice on DBA/2J (D2) and B6D2F1 (B6D2F1) backgrounds develop normally throughout gestation and do not develop dilated cardiomyopathy. However, the D2 mice do develop a severe metabolic acidosis and survive for only up to 12 d after birth. B6D2F1) mice have a milder form of metabolic acidosis and can survive for up to 3 weeks. The marked difference in lifespans and the development of dilated cardiomyopathy in the B6 but not the D2 or B6D2F1 backgrounds indicate the possible existence of genetic modifiers that provide protection to the developing hearts in the absence of MnSOD.


Assuntos
Acidose/metabolismo , Cardiomiopatia Dilatada/enzimologia , Morte Fetal/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Acidose/genética , Aconitato Hidratase/genética , Aconitato Hidratase/metabolismo , Animais , Cardiomiopatia Dilatada/genética , Catalase/genética , Catalase/metabolismo , Morte Fetal/genética , Genótipo , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Rim/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Camundongos , Camundongos Knockout , Camundongos Mutantes , Mitocôndrias/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Fenótipo , Superóxido Dismutase/deficiência , Regulação para Cima
7.
Free Radic Biol Med ; 31(8): 1018-30, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11595386

RESUMO

Manganese superoxide dismutase (MnSOD) is essential in protecting mitochondria against the damaging effects of superoxide radicals (O(2)(*-)), and increased expression of MnSOD protects cells and transgenic animals from various forms of oxidative stress. In addition, increased levels of MnSOD have been shown to slow down cell growth and induce differentiation. To study the effects of high MnSOD levels in vivo, we generated a series of transgenic mice using a mouse genomic sequence under control of the endogenous promoter. Four transgenic lines produced by pronuclear DNA injection exhibited up to 2-fold elevated MnSOD levels in brain and heart. However, using an embryonic stem cell approach, a line having 10-fold elevated MnSOD levels in the brain and 6- to 7-fold elevated levels in the heart and kidney was generated. Surprisingly, the genetic background of this transgenic line influenced the expression level of the transgene, with DBA/2 (D2) and C57BL/6 (B6) mice exhibiting low- and high-level transgene expression, respectively. This difference was the result of an increased transcription rate of the transgene. High-level MnSOD expression in B6 animals was associated with small size, male infertility, and decreased female fertility. These features are absent on the D2 background and indicate that high levels of MnSOD activity may interfere with normal growth and fertility.


Assuntos
Retardo do Crescimento Fetal/genética , Infertilidade/genética , Superóxido Dismutase/genética , Transcrição Gênica/genética , Transgenes/genética , Regulação para Cima/genética , Animais , Células da Medula Óssea/metabolismo , Encéfalo/metabolismo , Catalase/metabolismo , Feminino , Fibroblastos/metabolismo , Glutationa Redutase/metabolismo , Infertilidade/patologia , Células Intersticiais do Testículo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Miocárdio/metabolismo , Especificidade da Espécie , Superóxido Dismutase/metabolismo , Transferência Intratubária do Zigoto/métodos
8.
Am J Hum Genet ; 69(5): 1033-45, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11590545

RESUMO

May-Hegglin anomaly (MHA) and Fechtner (FTNS) and Sebastian (SBS) syndromes are autosomal dominant platelet disorders that share macrothrombocytopenia and characteristic leukocyte inclusions. FTNS has the additional clinical features of nephritis, deafness, and cataracts. Previously, mutations in the nonmuscle myosin heavy chain 9 gene (MYH9), which encodes nonmuscle myosin heavy chain IIA (MYHIIA), were identified in all three disorders. The spectrum of mutations and the genotype-phenotype and structure-function relationships in a large cohort of affected individuals (n=27) has now been examined. Moreover, it is demonstrated that MYH9 mutations also result in two other FTNS-like macrothrombocytopenia syndromes: Epstein syndrome (EPS) and Alport syndrome with macrothrombocytopenia (APSM). In all five disorders, MYH9 mutations were identified in 20/27 (74%) affected individuals. Four mutations, R702C, D1424N, E1841K, and R1933X, were most frequent. R702C and R702H mutations were only associated with FTNS, EPS, or APSM, thus defining a region of MYHIIA critical in the combined pathogenesis of macrothrombocytopenia, nephritis, and deafness. The E1841K, D1424N, and R1933X coiled-coil domain mutations were common to both MHA and FTNS. Haplotype analysis using three novel microsatellite markers revealed that three E1841K carriers--one with MHA and two with FTNS--shared a common haplotype around the MYH9 gene, suggesting a common ancestor. The two new globular-head mutations, K371N and R702H, as well as the recently identified MYH9 mutation, R705H, which results in DFNA17, were modeled on the basis of X-ray crystallographic data. Altogether, our data suggest that MHA, SBS, FTNS, EPS, and APSM comprise a phenotypic spectrum of disorders, all caused by MYH9 mutations. On the basis of our genetic analyses, the name "MYHIIA syndrome" is proposed to encompass all of these disorders.


Assuntos
Genes Dominantes/genética , Proteínas Motores Moleculares , Mutação/genética , Cadeias Pesadas de Miosina/genética , Miosina não Muscular Tipo IIA/genética , Trombocitopenia/genética , Sequência de Aminoácidos , Cromossomos/genética , Análise Mutacional de DNA , Evolução Molecular , Éxons/genética , Haplótipos/genética , Humanos , Repetições de Microssatélites/genética , Modelos Moleculares , Dados de Sequência Molecular , Cadeias Pesadas de Miosina/química , Nefrite Hereditária/genética , Nefrite Hereditária/fisiopatologia , Miosina não Muscular Tipo IIA/química , Fenótipo , Mapeamento Físico do Cromossomo , Conformação Proteica , Alinhamento de Sequência , Relação Estrutura-Atividade , Síndrome , Terminologia como Assunto , Trombocitopenia/fisiopatologia
9.
Genesis ; 30(4): 274-9, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11536434

RESUMO

Mice inheriting both copies of MMU12 either maternally or paternally demonstrate imprinting effects. Whereas maternal uniparental disomy 12 (matUPD12) fetuses are growth retarded and die perinatally, paternal UPD12 (patUPD12) fetuses die during late gestation and exhibit placentomegaly and skeletal muscle maturation defects. To examine further the developmental consequences of UPD12, we intercrossed mouse stocks heterozygous for Robertsonian translocation chromosomes (8.12) and (10.12). We report that at 13.5-14.5 dg patUPD12 hearts exhibit increased ventricular diameter, thinner, less compact myocardium, and deep intertrabecular recesses when compared to controls. These data provide evidence for cardiac failure, a lethal condition, and suggest a role for an imprinted gene(s) in normal heart development.


Assuntos
Cardiomiopatias/embriologia , Cardiomiopatias/genética , Aberrações Cromossômicas/genética , Impressão Genômica/genética , Miocárdio/patologia , Animais , Aberrações Cromossômicas/embriologia , Cruzamentos Genéticos , Feminino , Morte Fetal/genética , Coração/embriologia , Heterozigoto , Cariotipagem , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Translocação Genética/genética
10.
Proc Natl Acad Sci U S A ; 98(18): 10439-44, 2001 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-11504920

RESUMO

Age-related degeneration of basal forebrain cholinergic neurons (BFCNs) contributes to cognitive decline in Alzheimer's disease and Down's syndrome. With aging, the partial trisomy 16 (Ts65Dn) mouse model of Down's syndrome exhibited reductions in BFCN size and number and regressive changes in the hippocampal terminal fields of these neurons with respect to diploid controls. The changes were associated with significantly impaired retrograde transport of nerve growth factor (NGF) from the hippocampus to the basal forebrain. Intracerebroventricular NGF infusion reversed well established abnormalities in BFCN size and number and restored the deficit in cholinergic innervation. The findings are evidence that even BFCNs chronically deprived of endogenous NGF respond to an intervention that compensates for defective retrograde transport. We suggest that age-related cholinergic neurodegeneration may be a treatable disorder of failed retrograde NGF signaling.


Assuntos
Síndrome de Down/metabolismo , Fator de Crescimento Neural/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Transporte Biológico Ativo , Contagem de Células , Células , Fibras Colinérgicas/efeitos dos fármacos , Fibras Colinérgicas/metabolismo , Fibras Colinérgicas/patologia , Modelos Animais de Doenças , Síndrome de Down/tratamento farmacológico , Síndrome de Down/patologia , Hipocampo/metabolismo , Humanos , Infusões Parenterais , Camundongos , Camundongos Mutantes , Degeneração Neural/tratamento farmacológico , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Fator de Crescimento Neural/administração & dosagem , Fenótipo , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo , Prosencéfalo/patologia , Trissomia
11.
Am J Physiol Heart Circ Physiol ; 281(3): H1422-32, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11514315

RESUMO

Heart mitochondria from heterozygous (Sod2(-/+)) knockout mice have a 50% reduction in manganese superoxide dismutase (MnSOD) activity. The decrease in MnSOD activity was associated with increased mitochondrial oxidative damage as demonstrated by a decrease in the activities of iron sulfhydryl proteins sensitive to oxygen stress (aconitase and reduced nicotinamide adenine dinucleotide-oxidoreductase). Mitochondrial function was altered in the Sod2(-/+) mice, as shown by decreased respiration by complex I and an increase in the sensitivity of the permeability transition to induction by calcium and t-butylhydroperoxide. The increased induction of the permeability transition in heart mitochondria from Sod2(-/+.)mice was associated with increased release of cytochrome c and an increase in DNA fragmentation. Cardiomyocytes isolated from neonatal Sod2(-/+) and Sod2(-/-) mice were more sensitive to cell death than cardiomyocytes from Sod2(+/+) mice after t-butylhydroperoxide treatment, and this increased sensitivity was prevented by inhibiting the permeability transition with cyclosporin A. These experiments demonstrate that MnSOD may play an important role in the induction of the mitochondrial pathway of apoptosis in the heart, and this appears to occur primarily through the permeability transition.


Assuntos
Apoptose , Heterozigoto , Mitocôndrias Cardíacas/enzimologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Aconitato Hidratase/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citosol/enzimologia , Ativação Enzimática/fisiologia , Glutamato-Amônia Ligase/metabolismo , Glutationa/metabolismo , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/enzimologia , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/efeitos dos fármacos , Miocárdio/citologia , Miocárdio/enzimologia , Oxidantes/farmacologia , Estresse Oxidativo/fisiologia , Consumo de Oxigênio/fisiologia , Permeabilidade/efeitos dos fármacos , Superóxido Dismutase/deficiência , terc-Butil Hidroperóxido/farmacologia
12.
Arterioscler Thromb Vasc Biol ; 21(7): 1131-8, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11451741

RESUMO

Transgenic mice overexpressing Cu/Zn superoxide dismutase (hSod1Tg(+/0)) or catalase (hCatTg(+/0)) and knockout mice underexpressing manganese superoxide dismutase (Sod2(+/)(-)) or glutathione peroxidase-1 (Gpx1(-/-)) were used to study the effect of antioxidant enzymes on cell-mediated low density lipoprotein (LDL) oxidation and oxidized LDL (oxLDL)-induced apoptosis. Incubation of LDL with mouse aortic segments or smooth muscle cells (SMCs) resulted in a significant increase in LDL oxidation. However, LDL oxidation was significantly reduced when LDL was incubated with aortic segments and SMCs obtained from hSod1Tg(+/0) and hCatTg(+/0) mice compared with those obtained from wild-type mice. In contrast, LDL oxidation was significantly increased when LDL was incubated with aortic segments and SMCs obtained from Sod2(+/)(-) and Gpx1(-/-) mice. CuSO(4)-oxidized LDL increased DNA fragmentation and caspase activities in the primary cultures of mouse aortic SMCs. However, oxLDL-induced DNA fragmentation and caspase activities were reduced 50% in SMCs obtained from hSod1Tg(+/0) and hCatTg(+/0) mice compared with wild-type control mice. In contrast, oxLDL-induced DNA fragmentation and caspase activities were significantly increased in SMCs obtained from Sod2(+/)(-) and Gpx1(-/-) mice. These findings suggest that overexpression of Cu/Zn superoxide dismutase or catalase reduces cell-mediated LDL oxidation and oxLDL-induced apoptosis, whereas underexpression of manganese superoxide dismutase or glutathione peroxidase-1 increases cell-mediated LDL oxidation and oxLDL-induced apoptosis.


Assuntos
Aorta/citologia , Aorta/enzimologia , Apoptose , Lipoproteínas LDL/biossíntese , Lipoproteínas LDL/farmacologia , Músculo Liso Vascular/enzimologia , Animais , Antioxidantes/metabolismo , Caspases/metabolismo , Catalase/genética , Catalase/metabolismo , Catalase/fisiologia , Células Cultivadas , Técnicas de Cultura , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Glutationa Peroxidase/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Oxirredução , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase/fisiologia , Glutationa Peroxidase GPX1
13.
Audiol Neurootol ; 6(3): 117-23, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11474137

RESUMO

The participation of reactive oxygen species in aminoglycoside-induced ototoxicity has been deduced from observations that aminoglycoside-iron complexes catalyze the formation of superoxide radicals in vitro and that antioxidants attenuate ototoxicity in vivo. We therefore hypothesized that overexpression of Cu/Zn-superoxide dismutase (h-SOD1) should protect transgenic mice from ototoxicity. Immunocytochemistry confirmed expression of h-SOD1 in inner ear tissues of transgenic C57BL/6-TgN[SOD1]3Cje mice. Transgenic and nontransgenic littermates received kanamycin (400 mg/kg body weight/day) for 10 days beginning on day 10 after birth. Auditory thresholds were tested by evoked auditory brain stem responses at 1 month after birth. In nontransgenic animals, the threshold in the kanamycin-treated group was 45-50 dB higher than in saline-injected controls. In the transgenic group, kanamycin increased the threshold by only 15 dB over the respective controls. The effects were similar at 12 and 24 kHz. The protection by overexpression of superoxide dismutase supports the hypothesis that oxidant stress plays a significant role in aminoglycoside-induced ototoxicity. The results also suggest transgenic animals as suitable models to investigate the underlying mechanisms and possible strategies for prevention.


Assuntos
Antibacterianos/efeitos adversos , Cobre/metabolismo , Transtornos da Audição/induzido quimicamente , Transtornos da Audição/prevenção & controle , Canamicina/efeitos adversos , Superóxido Dismutase/metabolismo , Zinco/metabolismo , Animais , Antibacterianos/sangue , Limiar Auditivo/efeitos dos fármacos , Nitrogênio da Ureia Sanguínea , Cóclea/efeitos dos fármacos , Cóclea/metabolismo , Cóclea/patologia , Imuno-Histoquímica , Canamicina/sangue , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Vestíbulo do Labirinto/efeitos dos fármacos , Vestíbulo do Labirinto/metabolismo , Vestíbulo do Labirinto/patologia
14.
J Biol Chem ; 276(17): 14264-70, 2001 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-11297530

RESUMO

Manganese-superoxide dismutase (Sod2) removes mitochondrially derived superoxide (O(2)) at near-diffusion limiting rates and is the only antioxidant enzyme whose expression is regulated by numerous stimuli. Here it is shown that Sod2 also serves as a source of the intracellular signaling molecule H(2)O(2). Sod2-dependent increases in the steady-state levels of H(2)O(2) led to ERK1/2 activation and subsequent downstream transcriptional increases in matrix metalloproteinase-1 (MMP-1) expression, which were reversed by expression of the H(2)O(2)-detoxifying enzyme, catalase. In addition, a single nucleotide polymorphism has recently been identified (1G/2G) at base pair--1607 that creates an Ets site adjacent to an AP-1 site at base pair --1602 and has been shown to dramatically enhance transcription of the MMP-1 promoter. Luciferase promoter constructs containing either the 1G or 2G variation were 25- or 1000-fold more active when transiently transfected into Sod2-overexpressing cell lines, respectively. The levels of MMP-2, -3, and -7 were also increased in the Sod2-overexpressing cell lines, suggesting that Sod2 may function as a "global" redox regulator of MMP expression. In addition, Sod2(-/+) mouse embryonic fibroblasts failed to respond to the cytokine-mediated induction of the murine functional analog of MMP-1, MMP-13. This study provides evidence that the modulation of Sod2 activity by a wide array of pathogenic and inflammatory stimuli may be utilized by the cell as a primary signaling mechanism leading to matrix metalloproteinase expression.


Assuntos
Peróxido de Hidrogênio/farmacologia , Metaloproteinases da Matriz/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Superóxido Dismutase/metabolismo , Animais , Northern Blotting , Western Blotting , Catalase/genética , Catalase/metabolismo , Separação Celular , Colagenases/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Fibroblastos/metabolismo , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica , Humanos , Imidazóis/farmacologia , Interleucina-1/genética , Luciferases/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 13 da Matriz , Camundongos , Proteína Quinase 3 Ativada por Mitógeno , Modelos Biológicos , Oxirredução , Fosforilação , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Piridinas/farmacologia , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Superóxido Dismutase/genética , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas
15.
J Exp Med ; 193(8): 925-34, 2001 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-11304553

RESUMO

Manganese superoxide dismutase 2 (SOD2) is a critical component of the mitochondrial pathway for detoxification of O2(-), and targeted disruption of this locus leads to embryonic or neonatal lethality in mice. To follow the effects of SOD2 deficiency in cells over a longer time course, we created hematopoietic chimeras in which all blood cells are derived from fetal liver stem cells of Sod2 knockout, heterozygous, or wild-type littermates. Stem cells of each genotype efficiently rescued hematopoiesis and allowed long-term survival of lethally irradiated host animals. Peripheral blood analysis of leukocyte populations revealed no differences in reconstitution kinetics of T cells, B cells, or myeloid cells when comparing Sod2(+/+), Sod2(-/-), and Sod2(+/-) fetal liver recipients. However, animals receiving Sod2(-/-) cells were persistently anemic, with findings suggestive of a hemolytic process. Loss of SOD2 in erythroid progenitor cells results in enhanced protein oxidative damage, altered membrane deformation, and reduced survival of red cells. Treatment of anemic animals with Euk-8, a catalytic antioxidant with both SOD and catalase activities, significantly corrected this oxidative stress-induced condition. Such therapy may prove useful in treatment of human disorders such as sideroblastic anemia, which SOD2 deficiency most closely resembles.


Assuntos
Anemia Hemolítica/tratamento farmacológico , Anemia Hemolítica/genética , Antioxidantes/uso terapêutico , Etilenodiaminas/uso terapêutico , Mitocôndrias/enzimologia , Compostos Organometálicos/uso terapêutico , Superóxido Dismutase/deficiência , Anemia Hemolítica/sangue , Animais , Linfócitos B/imunologia , Células da Medula Óssea/imunologia , Transplante de Células , Transplante de Tecido Fetal/fisiologia , Sequestradores de Radicais Livres/uso terapêutico , Genótipo , Heterozigoto , Isoenzimas/deficiência , Isoenzimas/genética , Fígado/citologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Baço/imunologia , Superóxido Dismutase/genética , Linfócitos T/imunologia , Timo/imunologia , Quimeras de Transplante
16.
Cancer Res ; 61(8): 3472-9, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11309310

RESUMO

Mucinous colorectal cancers exhibit a characteristic set of molecular genetic alterations and may be derived from progenitor cells committed to the goblet cell lineage. Previously, we demonstrated that the MUC2 mucin gene promoter drives transgene reporter expression with high specificity in small intestinal goblet cells of transgenic mice. On the basis of these experiments, we reasoned that the MUC2 promoter could be used to drive SV40 T antigen (Tag) expression in the same cell type, decoupling them from their normal antiproliferative controls. A line of mice was established (MUCTag6) that expressed Tag in intestinal goblet cells as determined by RNA blot and immunohistochemical analysis. These goblet cells were markedly involuted however, most notably in the villi. Endogenous intestinal MUC2 message levels were reduced to about one third the normal level in these mice. However, absorptive cell lineage markers were comparable with nontransgenics. Bromodeoxyuridine-positive S-phase cells are limited to crypts in nontransgenic intestine but are present in both crypts and villi in MUCTag6. In contrast, mitotic cells were not present in the villi, indicating that MUCTag6 villi goblet cells do not progress into M phase. Apoptotic cells positive for terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling were increased more than fourfold in MUCTag6 villi (P < 0.0001), and apoptotic goblet cells were evident. Electron microscopic examination of MUCTag6 intestinal villi revealed the presence of degraded cell remnants containing mucin goblets together with other cell debris, further indicating apoptosis of the goblet cell lineage. Thus, the expression of Tag in intestinal goblet cells releases them from normal antiproliferative controls, causing their inappropriate entry into S phase even after they transverse the crypt/villus junction. They do not, however, progress to M phase. Instead, they undergo apoptosis with a high degree of efficiency in S or G(2) phase. These experiments demonstrate that apoptosis effectively blocks inappropriate goblet cell proliferation in the intestine, supporting its proposed role as an antineoplastic mechanism.


Assuntos
Antígenos Transformantes de Poliomavirus/biossíntese , Apoptose/fisiologia , Movimento Celular/fisiologia , Células Caliciformes/citologia , Intestino Delgado/citologia , Mucinas/genética , Animais , Antígenos Transformantes de Poliomavirus/genética , Feminino , Células Caliciformes/imunologia , Células Caliciformes/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Microvilosidades/fisiologia , Mucina-2 , Oncogenes , Regiões Promotoras Genéticas , Fase S/fisiologia
17.
Int J Cancer ; 96(1): 11-21, 2001 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-11241326

RESUMO

Intratracheal (IT) injection of the transgene for human manganese superoxide dismutase in plasmid/liposome (SOD2-PL) complex prior to irradiation protects C57BL/6J mice from whole lung irradiation-induced organizing alveolitis/fibrosis. Transgene mRNA was detected in alveolar type II (AT-II) and tracheobronchial tree cells explanted to culture 48 hours after gene therapy. To determine whether constitutive overexpression of murine MnSOD (Sod2) in whole lung or surfactant promoter-restricted AT-II cells (SP1)-SOD2 mice would provide intrinsic radioresistance, transgenic mice of two strains were compared with age-matched controls. Other groups of surfactant promoter-restricted (SP1)-SOD2 transgenic mice or control FeVB/NHsd mice received IT SOD2-PL gene therapy prior to irradiation. There was no significant intrinsic lung protection in either strain of MnSOD transgenic mice. The SP1-SOD2 transgenic mice were protected from lung damage by IT injection of the human SOD2-PL complex 24 hours prior to irradiation. Thus, overexpression of either human SOD2 or murine Sod2 in the lungs of transgenic mice does not provide intrinsic lung irradiation protection. The overexpression of SOD2 in the SP1-SOD2 mice may have made the mice more sensitive to irradiation.


Assuntos
Pulmão/enzimologia , Superóxido Dismutase/biossíntese , Animais , Brônquios/citologia , Brônquios/efeitos da radiação , Células Cultivadas , Relação Dose-Resposta à Radiação , Fibrose/etiologia , Terapia Genética , Humanos , Lipossomos/metabolismo , Pulmão/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Alvéolos Pulmonares/efeitos da radiação , RNA Mensageiro/metabolismo , Tolerância a Radiação/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Superóxido Dismutase/genética , Tensoativos/metabolismo , Fatores de Tempo , Traqueia/citologia , Traqueia/efeitos da radiação
18.
Exp Neurol ; 167(1): 189-95, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11161607

RESUMO

There is substantial evidence implicating mitochondrial dysfunction and free radical generation as major mechanisms of neuronal death in neurodegenerative diseases. The major free radical scavenging enzyme in mitochondria is manganese superoxide dismutase (SOD2). In the present study we investigated the susceptibility of mice with a partial deficiency of SOD2 to the neurotoxins 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine (MPTP), 3-nitropropionic acid (3-NP), and malonate, which are commonly used animal models of Parkinson's and Huntington's disease. Heterozygous SOD2 knockout (SOD2(+/-)) mice showed no evidence of neuropathological or behavioral abnormalities at 2-4 months of age. Compared to littermate wild-type mice, mice with partial SOD2 deficiency showed increased vulnerability to dopamine depletion after systemic MPTP treatment and significantly larger striatal lesions produced by both 3-NP and malonate. SOD2(+/-) mice also showed an increased production of "hydroxyl" radicals after malonate injection measured with the salicylate hydroxyl radical trapping method. These results provide further evidence that reactive oxygen species play an important role in the neurotoxicity of MPTP, malonate, and 3-NP. These findings show that a subclinical deficiency in a free radical scavenging enzyme may act in concert with environmental toxins to produce selective neurodegeneration.


Assuntos
Predisposição Genética para Doença , Gentisatos , Glicoproteínas de Membrana , Proteínas de Membrana Transportadoras , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso , Doenças Neurodegenerativas/metabolismo , Neurotoxinas/metabolismo , Superóxido Dismutase/deficiência , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Proteínas de Transporte/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina , Feminino , Predisposição Genética para Doença/genética , Heterozigoto , Ácido Homovanílico/metabolismo , Hidroxibenzoatos/metabolismo , Radical Hidroxila/metabolismo , Masculino , Malonatos , Camundongos , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Doenças Neurodegenerativas/induzido quimicamente , Doenças Neurodegenerativas/patologia , Neurotoxinas/toxicidade , Nitrocompostos , Propionatos , Ácido Salicílico/metabolismo , Superóxido Dismutase/genética
19.
J Biol Chem ; 276(4): 2775-9, 2001 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-11035038

RESUMO

Hereditary non-X-linked nephrogenic diabetes insipidus (NDI) is caused by mutations in the aquaporin-2 (AQP2) water channel. In transfected cells, the human disease-causing mutant AQP2-T126M is retained at the endoplasmic reticulum (ER) where it is functional and targetable to the plasma membrane with chemical chaperones. A mouse knock-in model of NDI was generated by targeted gene replacement using a Cre-loxP strategy. Along with T126M, mutations H122S, N124S, and A125T were introduced to preserve the consensus sequence for N-linked glycosylation found in human AQP2. Breeding of heterozygous mice yielded the expected Mendelian distribution with 26 homozygous mutant offspring of 99 live births. The mutant mice appeared normal at 2-3 days after birth but failed to thrive and generally died by day 6 if not given supplemental fluid. Urine/serum analysis showed a urinary concentrating defect with serum hyperosmolality and low urine osmolality that was not increased by a V2 vasopressin agonist. Northern blot analysis showed up-regulated AQP2-T126M transcripts of identical size to wild-type AQP2. Immunoblots showed complex glycosylation of wild-type AQP2 but mainly endoglycosidase H-sensitive core glycosylation of AQP2-T126M indicating ER-retention. Biochemical analysis revealed that the AQP2-T126M protein was resistant to detergent solubilization. Kidneys from mutant mice showed collecting duct dilatation, papillary atrophy, and unexpectedly, some plasma membrane AQP2 staining. The severe phenotype of the AQP2 mutant mice compared with that of mice lacking kidney water channels AQP1, AQP3, and AQP4 indicates a critical role for AQP2 in neonatal renal function in mice. Our results establish a mouse model of human autosomal NDI and provide the first in vivo biochemical data on a disease-causing AQP2 mutant.


Assuntos
Aquaporinas/genética , Diabetes Insípido Nefrogênico/genética , Modelos Animais de Doenças , Genes Recessivos , Camundongos Transgênicos , Animais , Animais Recém-Nascidos , Aquaporina 2 , Aquaporina 6 , Aquaporinas/biossíntese , Diabetes Insípido Nefrogênico/mortalidade , Células Epiteliais/patologia , Túbulos Renais Coletores/patologia , Camundongos
20.
Blood ; 96(10): 3447-51, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11071640

RESUMO

Families with 3 different syndromes characterized by autosomal dominant inheritance of low platelet count and giant platelets were studied. Fechtner syndrome is an autosomal-dominant variant of Alport syndrome manifested by nephritis, sensorineural hearing loss, and cataract formation in addition to macrothrombocytopenia and polymorphonuclear inclusion bodies. Sebastian platelet syndrome is an autosomal-dominant macrothrombocytopenia combined with neutrophil inclusions that differ from those found in May-Hegglin syndrome or Chediak-Higashi syndrome or the Dohle bodies described in patients with sepsis. These inclusions are, however, similar to those described in Fechtner syndrome. Other features of Alport syndrome, though, including deafness, cataracts, and nephritis, are absent in Sebastian platelet syndrome. Epstein syndrome is characterized by macrothrombocytopenia without neutrophil inclusions, in addition to the classical Alport manifestations-deafness, cataracts, and nephritis-and it is also inherited in an autosomal-dominant mode. We mapped the disease-causing gene to the long arm of chromosome 22 in an Italian family with Fechtner syndrome, 2 German families with the Sebastian platelet syndrome, and an American family with the Epstein syndrome. Four markers on chromosome 22q yielded an LOD score greater than 2.76. A maximal 2-point LOD score of 3.41 was obtained with the marker D22S683 at a recombination fraction of 0.00. Recombination analysis placed the disease-causing gene in a 3.37-Mb interval between the markers D22S284 and D22S693. The disease-causing gene interval in these 3 syndromes is similar to the interval described recently in an Israeli family with a slightly different Fechtner syndrome than the one described here. Recombination analysis of these 3 syndromes refines the interval containing the disease-causing gene from 5.5 Mb to 3.37 Mb. The clinical likeness and the similar interval containing the disease-causing gene suggest that the 3 different syndromes may arise from a similar genetic defect.


Assuntos
Síndrome de Bernard-Soulier/genética , Cromossomos Humanos Par 22 , Transtornos Plaquetários/genética , Mapeamento Cromossômico , Saúde da Família , Feminino , Genes Dominantes , Ligação Genética , Marcadores Genéticos , Genótipo , Humanos , Escore Lod , Masculino , Linhagem , Trombocitopenia/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...