Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
FASEB J ; 34(12): 16224-16242, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33058343

RESUMO

Importin α proteins play a central role in the transport of cargo from the cytoplasm to the nucleus. In this study, we observed that male knock-out mice for importin α4, which is encoded by the Kpna4 gene (Kpna4-/- ), were subfertile and yielded smaller litter sizes than those of wild-type (WT) males. In contrast, mice lacking the closely related importin α3 (Kpna3-/- ) were fertile. In vitro fertilization and sperm motility assays demonstrated that sperm from Kpna4-/- mice had significantly reduced quality and motility. In addition, acrosome reaction was also impaired in Kpna4-/- mice. Transmission electron microscopy revealed striking defects, including abnormal head morphology and multiple axoneme structures in the flagella of Kpna4-/- mice. A five-fold increase in the frequency of abnormalities in Kpna4-/- mice compared to WT mice indicates the functional importance of importin α4 in normal sperm development. Moreover, Nesprin-2, which is a component of the linker of nucleus and cytoskeleton complex, was expressed at lower levels in sperm from Kpna4-/- mice and was localized with abnormal axonemes, suggesting incorrect formation of the nuclear membrane-cytoskeleton structure during spermiogenesis. Proteomics analysis of Kpna4-/- testis showed significantly altered expression of proteins related to sperm formation, which provided evidence that genetic loss of importin α4 perturbed chromatin status. Collectively, these findings indicate that importin α4 is critical for establishing normal sperm morphology in mice, providing new insights into male germ cell development by highlighting the requirement of importin α4 for normal fertility.


Assuntos
Fertilidade/genética , Infertilidade Masculina/genética , Carioferinas/genética , Motilidade dos Espermatozoides/genética , Espermatozoides/anormalidades , alfa Carioferinas/genética , Reação Acrossômica/genética , Animais , Flagelos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espermatogênese/genética , Testículo/anormalidades
2.
Biosci Biotechnol Biochem ; 80(2): 288-90, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26745679
3.
J Lipid Res ; 56(12): 2286-96, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26438561

RESUMO

Exercise training influences phospholipid fatty acid composition in skeletal muscle and these changes are associated with physiological phenotypes; however, the molecular mechanism of this influence on compositional changes is poorly understood. Peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), a nuclear receptor coactivator, promotes mitochondrial biogenesis, the fiber-type switch to oxidative fibers, and angiogenesis in skeletal muscle. Because exercise training induces these adaptations, together with increased PGC-1α, PGC-1α may contribute to the exercise-mediated change in phospholipid fatty acid composition. To determine the role of PGC-1α, we performed lipidomic analyses of skeletal muscle from genetically modified mice that overexpress PGC-1α in skeletal muscle or that carry KO alleles of PGC-1α. We found that PGC-1α affected lipid profiles in skeletal muscle and increased several phospholipid species in glycolytic muscle, namely phosphatidylcholine (PC) (18:0/22:6) and phosphatidylethanolamine (PE) (18:0/22:6). We also found that exercise training increased PC (18:0/22:6) and PE (18:0/22:6) in glycolytic muscle and that PGC-1α was required for these alterations. Because phospholipid fatty acid composition influences cell permeability and receptor stability at the cell membrane, these phospholipids may contribute to exercise training-mediated functional changes in the skeletal muscle.


Assuntos
Músculo Esquelético/metabolismo , Fosfolipídeos/metabolismo , Condicionamento Físico Animal/fisiologia , Fatores de Transcrição/metabolismo , Animais , Humanos , Masculino , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/genética
4.
Biochem J ; 456(3): 373-83, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24094090

RESUMO

Fabry disease is a lysosomal storage disorder in which neutral glycosphingolipids, predominantly Gb3 (globotriaosylceramide), accumulate due to deficient α-Gal A (α-galactosidase A) activity. The GLAko (α-Gal A-knockout) mouse has been used as a model for Fabry disease, but it does not have any symptomatic abnormalities. In the present study, we generated a symptomatic mouse model (G3Stg/GLAko) by cross-breeding GLAko mice with transgenic mice expressing human Gb3 synthase. G3Stg/GLAko mice had high Gb3 levels in major organs, and their serum Gb3 level at 5-25 weeks of age was 6-10-fold higher than that in GLAko mice of the same age. G3Stg/GLAko mice showed progressive renal impairment, with albuminuria at 3 weeks of age, decreased urine osmolality at 5 weeks, polyuria at 10 weeks and increased blood urea nitrogen at 15 weeks. The urine volume and urinary albumin concentration were significantly reduced in the G3Stg/GLAko mice when human recombinant α-Gal A was administered intravenously. These data suggest that Gb3 accumulation is a primary pathogenic factor in the symptomatic phenotype of G3Stg/GLAko mice, and that this mouse line is suitable for studying the pathogenesis of Fabry disease and for preclinical studies of candidate therapies.


Assuntos
Doença de Fabry/metabolismo , Galactosiltransferases/metabolismo , Triexosilceramidas/biossíntese , alfa-Galactosidase/farmacologia , Albuminúria/tratamento farmacológico , Albuminúria/genética , Albuminúria/metabolismo , Albuminúria/patologia , Animais , Modelos Animais de Doenças , Doença de Fabry/tratamento farmacológico , Doença de Fabry/genética , Doença de Fabry/patologia , Galactosiltransferases/genética , Humanos , Camundongos , Camundongos Transgênicos , Triexosilceramidas/genética , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo
5.
PLoS Genet ; 7(10): e1002279, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21998593

RESUMO

Upon fertilization, reprogramming of gene expression is required for embryo development. This step is marked by DNA demethylation and changes in histone variant composition. However, little is known about the molecular mechanisms causing these changes and their impact on histone modifications. We examined the global deposition of the DNA replication-dependent histone H3.1 and H3.2 variants and the DNA replication-independent H3.3 variant after fertilization in mice. We showed that H3.3, a euchromatic marker of gene activity, transiently disappears from the maternal genome, suggesting erasure of the oocyte-specific modifications carried by H3.3. After fertilization, H3.2 is incorporated into the transcriptionally silent heterochromatin, whereas H3.1 and H3.3 occupy unusual heterochromatic and euchromatin locations, respectively. After the two-cell stage, H3.1 and H3.3 variants resume their usual respective locations on heterochromatin and euchromatin. Preventing the incorporation of H3.1 and H3.2 by knockdown of the histone chaperone CAF-1 induces a reciprocal increase in H3.3 deposition and impairs heterochromatin formation. We propose that the deposition of different H3 variants influences the functional organization of chromatin. Taken together, these findings suggest that dynamic changes in the deposition of H3 variants are critical for chromatin reorganization during epigenetic reprogramming.


Assuntos
Cromatina/genética , Eucromatina/genética , Histonas/genética , Histonas/metabolismo , Oogênese/genética , Proteínas/genética , Proteínas/metabolismo , Animais , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , Epigênese Genética , Eucromatina/metabolismo , Exorribonucleases , Feminino , Fertilização/genética , Desenvolvimento Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Heterocromatina/genética , Heterocromatina/metabolismo , Camundongos , Camundongos Transgênicos , Células NIH 3T3 , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Repressoras , Ribonucleases
6.
Nat Med ; 12(4): 466-72, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16550188

RESUMO

Adult T-cell leukemia-lymphoma (ATLL) is a group of T-cell malignancies caused by infection with human T-lymphotropic virus type I (HTLV-I). Although the pathogenesis of ATLL remains incompletely understood, the viral regulatory protein Tax is centrally involved in cellular transformation. Here we describe the generation of HTLV-I Tax transgenic mice using the Lck proximal promoter to restrict transgene expression to developing thymocytes. After prolonged latency periods, transgenic mice developed diffuse large-cell lymphomas and leukemia with clinical, pathological and immunological features characteristic of acute ATLL. Transgenic mice were functionally immunocompromised and they developed opportunistic infections. Fulminant disease also developed rapidly in SCID mice after engraftment of lymphomatous cells from transgenic mice. Flow cytometry showed that the cells were CD4(-) and CD8(-), but CD44(+), CD25(+) and cytoplasmic CD3(+). This phenotype is indicative of a thymus-derived pre-T-cell phenotype, and disease development was associated with the constitutive activation of NF-kappaB. Our model accurately reproduces human disease and will provide a tool for analysis of the molecular events in transformation and for the development of new therapeutics.


Assuntos
Genes pX , Vírus Linfotrópico T Tipo 1 Humano/genética , Leucemia Linfoide/patologia , Neoplasias do Timo/patologia , Animais , Biomarcadores , Complexo CD3/imunologia , Complexo CD3/metabolismo , Mapeamento Cromossômico , Cromossomos , Modelos Animais de Doenças , Ensaio de Desvio de Mobilidade Eletroforética , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Vírus Linfotrópico T Tipo 1 Humano/ultraestrutura , Humanos , Imuno-Histoquímica , Leucemia Linfoide/genética , Leucemia Linfoide/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Camundongos Transgênicos , Transplante de Neoplasias , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias do Timo/imunologia , Transgenes , Transplante Homólogo
7.
Mol Genet Metab ; 106(1): 92-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22436580

RESUMO

Therapeutic chaperone effect of a valienamine derivative N-octyl 4-epi-ß-valienamine (NOEV) was studied in G(M1)-gangliosidosis model mice. Phamacokinetic analysis revealed rapid intestinal absorption and renal excretion after oral administration. Intracellular accumulation was not observed after continuous treatment. NOEV was delivered to the central nervous system through the blood-brain barrier to induce high expression of the apparently deficient ß-galactosidase activity. NOEV treatment starting at the early stage of disease resulted in remarkable arrest of neurological progression within a few months. Survival time was significantly prolonged. This result suggests that NOEV chaperone therapy will be clinically effective for prevention of neuronal damage if started early in life hopefully also in human patients with G(M1)-gangliosidosis.


Assuntos
Gangliosidose GM1/terapia , Hexosaminas/administração & dosagem , Chaperonas Moleculares/administração & dosagem , beta-Galactosidase/genética , beta-Glucosidase/genética , Animais , Barreira Hematoencefálica , Sistema Nervoso Central/enzimologia , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Gangliosidose GM1/enzimologia , Gangliosidose GM1/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hexosaminas/farmacocinética , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Chaperonas Moleculares/farmacocinética , Urinálise
8.
Hum Mutat ; 32(7): 843-52, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21520340

RESUMO

ß-Galactosidase deficiency is a group of lysosomal lipid storage disorders with an autosomal recessive trait. It causes two clinically different diseases, G(M1) -gangliosidosis and Morquio B disease. It is caused by heterogeneous mutations in the GLB1 gene coding for the lysosomal acid ß-galactosidase. We have previously reported the chaperone effect of N-octyl-4-epi-ß-valienamine (NOEV) on mutant ß-galactosidase proteins. In this study, we performed genotype analyses of patients with ß-galactosidase deficiency and identified 46 mutation alleles including 9 novel mutations. We then examined the NOEV effect on mutant ß-galactosidase proteins by using six strains of patient-derived skin fibroblast. We also performed mutagenesis to identify ß-galactosidase mutants that were responsive to NOEV and found that 22 out of 94 mutants were responsive. Computational structural analysis revealed the mode of interaction between human ß-galactosidase and NOEV. Moreover, we confirmed that NOEV reduced G(M1) accumulation and ameliorated the impairments of lipid trafficking and protein degradation in ß-galactosidase deficient cells. These results provided further evidence to NOEV as a promising chaperone compound for ß-galactosidase deficiency.


Assuntos
Fibroblastos/efeitos dos fármacos , Gangliosidose GM1/tratamento farmacológico , Hexosaminas/farmacologia , beta-Galactosidase/química , beta-Galactosidase/metabolismo , Animais , Células Cultivadas , Estabilidade Enzimática , Fibroblastos/enzimologia , Gangliosidose GM1/enzimologia , Expressão Gênica , Vetores Genéticos , Hexosaminas/química , Hexosaminas/uso terapêutico , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucopolissacaridose IV/genética , Mutação de Sentido Incorreto/genética , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , beta-Galactosidase/genética
9.
J Neurochem ; 118(3): 399-406, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21574998

RESUMO

G(M1) -gangliosidosis is a fatal neurodegenerative disorder caused by deficiency of lysosomal acid ß-galactosidase (ß-gal). Accumulation of its substrate ganglioside G(M1) (G(M1) ) in lysosomes and other parts of the cell leads to progressive neurodegeneration, but underlying mechanisms remain unclear. Previous studies demonstrated an essential role for interaction of G(M1) with tropomyosin receptor kinase (Trk) receptors in neuronal growth, survival and differentiation. In this study we demonstrate accumulation of G(M1) in the cell-surface rafts and lysosomes of the ß-gal knockout (ß-gal-/-) mouse brain association with accumulation of Trk receptors and enhancement of its downstream signaling. Immunofluorescence and subcellular fractionation analysis revealed accumulation of Trk receptors in the late endosomes/lysosomes of the ß-gal-/- mouse brain and their association with ubiquitin and p62. Administration of a chemical chaperone to ß-gal-/- mouse expressing human mutant R201C protein resulted in a marked reduction of intracellular storage of G(M1) and phosphorylated Trk. These findings indicate that G(M1) accumulation in rafts causes activation of Trk signaling, which may participate in the pathogenesis of G(M1) -gangliosidosis.


Assuntos
Química Encefálica/fisiologia , Gangliosidose GM1/metabolismo , Hexosaminas/farmacologia , Lisossomos/metabolismo , Chaperonas Moleculares/farmacologia , Receptor trkA/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Encéfalo/patologia , Química Encefálica/efeitos dos fármacos , Células Cultivadas , Imunofluorescência , Humanos , Imunoprecipitação , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Crescimento Neural/fisiologia , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , beta-Galactosidase/genética , beta-Galactosidase/farmacologia
10.
Circ Res ; 105(1): 25-32, 2009 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-19478204

RESUMO

Obese adipose tissue is markedly infiltrated by macrophages, suggesting that they may participate in the inflammatory pathways that are activated in obese adipose tissue. Evidence has suggested that saturated fatty acids released via adipocyte lipolysis serve as a naturally occurring ligand that stimulates Toll-like receptor (TLR)4 signaling, thereby inducing the inflammatory responses in macrophages in obese adipose tissue. Through a combination of cDNA microarray analyses of saturated fatty acid-stimulated macrophages in vitro and obese adipose tissue in vivo, here we identified activating transcription factor (ATF)3, a member of the ATF/cAMP response element-binding protein family of basic leucine zipper-type transcription factors, as a target gene of saturated fatty acids/TLR4 signaling in macrophages in obese adipose tissue. Importantly, ATF3, when induced by saturated fatty acids, can transcriptionally repress tumor necrosis factor-alpha production in macrophages in vitro. Chromatin immunoprecipitation assay revealed that ATF3 is recruited to the region containing the activator protein-1 site of the endogenous tumor necrosis factor-alpha promoter. Furthermore, transgenic overexpression of ATF3 specifically in macrophages results in the marked attenuation of proinflammatory M1 macrophage activation in the adipose tissue from genetically obese KKA(y) mice fed high-fat diet. This study provides evidence that ATF3, which is induced in obese adipose tissue, acts as a transcriptional repressor of saturated fatty acids/TLR4 signaling, thereby revealing the negative feedback mechanism that attenuates obesity-induced macrophage activation. Our data also suggest that activation of ATF3 in macrophages offers a novel therapeutic strategy to prevent or treat obesity-induced adipose tissue inflammation.


Assuntos
Fator 3 Ativador da Transcrição/fisiologia , Tecido Adiposo/metabolismo , Ácidos Graxos/metabolismo , Ativação de Macrófagos , Obesidade/patologia , Receptor 4 Toll-Like/metabolismo , Animais , Linhagem Celular , Retroalimentação Fisiológica , Perfilação da Expressão Gênica , Inflamação , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos , Transdução de Sinais , Fatores de Transcrição
11.
Artigo em Inglês | MEDLINE | ID: mdl-21986317

RESUMO

Sugar chain abnormalities in glycolipids and glycoproteins are associated with various diseases. Here, we report an adult onset cardiac dilatation in a transgenic mouse line with Galß1,3GalNAc α2,3-sialyltransferase II (ST3Gal-II) transgenes. The transgenic hearts at the end-stage, at around 7 months old, were enlarged, with enlarged cavities and thin, low-tensile walls, typical of dilated cardiomyopathy. Although no apparent change was found in heart gangliosides, glycosylation of heart proteins was altered. Interestingly, sugar moieties not directly related to the ST3Gal-II catalytic reaction were also changed. Significant increases in calreticulin and calnexin were observed in hearts of the transgenic mice. These results suggest that expression of ST3Gal-II transgenes induces abnormal protein glycosylation, which disorganizes the endoplasmic/sarcoplasmic reticulum quality control system and elevates the calreticulin/calnexin level, resulting in suppression of cardiac function. The transgenic mice showed 100% incidence of adult onset cardiac dilatation, suggesting great potential as a new model for dilated cardiomyopathy.


Assuntos
Envelhecimento/patologia , Cardiomiopatia Dilatada/enzimologia , Cardiomiopatia Dilatada/patologia , Sialiltransferases/metabolismo , Transgenes/genética , Animais , Calnexina/metabolismo , Calreticulina/metabolismo , Modelos Animais de Doenças , Secções Congeladas , Gangliosídeos/metabolismo , Homozigoto , Lectinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocárdio/metabolismo , Miocárdio/patologia , Especificidade de Órgãos , Coloração e Rotulagem , beta-Galactosídeo alfa-2,3-Sialiltransferase
12.
Dev Biol ; 331(2): 140-51, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19409883

RESUMO

The neocortex and the hippocampus comprise several specific layers containing distinct neurons that originate from progenitors at specific development times, under the control of an adequate cell-division patterning mechanism. Although many molecules are known to regulate this cell-division patterning process, its details are not well understood. Here, we show that, in the developing cerebral cortex, the RP58 transcription repressor protein was expressed both in postmitotic glutamatergic projection neurons and in their progenitor cells, but not in GABAergic interneurons. Targeted deletion of the RP58 gene led to dysplasia of the neocortex and of the hippocampus, reduction of the number of mature cortical neurons, and defects of laminar organization, which reflect abnormal neuronal migration within the cortical plate. We demonstrate an impairment of the cell-division patterning during the late embryonic stage and an enhancement of apoptosis of the postmitotic neurons in the RP58-deficient cortex. These results suggest that RP58 controls cell division of progenitor cells and regulates the survival of postmitotic cortical neurons.


Assuntos
Diferenciação Celular/fisiologia , Córtex Cerebral/embriologia , Hipocampo/embriologia , Neurogênese/fisiologia , Neurônios/citologia , Proteínas Repressoras/fisiologia , Sequência de Aminoácidos , Animais , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Hipocampo/citologia , Hipocampo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Neurônios/fisiologia , Proteínas Repressoras/genética , Células-Tronco/citologia , Células-Tronco/fisiologia
13.
Drug Des Devel Ther ; 13: 555-568, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30787596

RESUMO

BACKGROUND: Anti-neutrophil cytoplasmic autoantibodies (ANCA) associated vasculitis is a pauci-immune disease with the inflammation of the small blood vessels. The efficacies of antibody drugs for induction therapies of vasculitis vary among cases. Here, we developed a novel clone of a single chain Fv region (ScFv) with vasculitis-specific therapeutic potential. MATERIALS AND METHODS: The clone, termed VasSF, was selected from our Escherichia coli expression library of recombinant human ScFv based on the therapeutic efficacy in an SCG/Kj mouse model of MPO-ANCA-associated vasculitis (MAAV), such as improvement of the urinary score and decreased crescent formation in glomeruli, granulomatous in lung, MPO-ANCA biomarkers, the anti-moesin antibody, and some cytokine levels. RESULTS: We identified vasculitis-associated apolipoprotein A-II (VAP2) as a target molecule of the clone and confirmed the independently-established VAP2 antibodies were also therapeutic in SCG/Kj mice. In MAAV, MPO-ANCA and cytokines stimulate neutrophils by facilitating heterodimer formation of VAP2 with apolipoprotein A-I in HDL. CONCLUSION: VasSF would constitute a novel antibody drug for vasculitis by suppressing the heterodimer formation of the apolipoproteins.


Assuntos
Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/tratamento farmacológico , Anticorpos de Cadeia Única/uso terapêutico , Animais , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/patologia , Relação Dose-Resposta a Droga , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Biblioteca de Peptídeos , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/uso terapêutico , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/isolamento & purificação
14.
Biochem Biophys Res Commun ; 367(3): 616-22, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18190792

RESUMO

G(M1)-gangliosidosis is an autosomal recessive lysosomal lipid storage disorder, caused by mutations of the lysosomal beta-galactosidase (beta-gal) and results in the accumulation of G(M1). The underlying mechanisms of neurodegeneration are poorly understood. Here we demonstrate increased autophagy in beta-gal-deficient (beta-gal(-/-)) mouse brains as evidenced by elevation of LC3-II and beclin-1 levels. Activation of autophagy in the beta-gal(-/-) brain was found to be accompanied with enhanced Akt-mTOR and Erk signaling. In addition, the mitochondrial cytochrome c oxidase activity was significantly decreased in brains and cultured astrocytes from beta-gal(-/-) mouse. Mitochondria isolated from beta-gal(-/-) astrocytes were morphologically abnormal and had a decreased membrane potential. These cells were more sensitive to oxidative stress than wild type cells and this sensitivity was suppressed by ATP, an autophagy inhibitor 3-methyladenine and a pan-caspase inhibitor z-VAD-fmk. These results suggest activation of autophagy leading to mitochondrial dysfunction in the brain of G(M1)-gangliosidosis.


Assuntos
Autofagia , Encéfalo/patologia , Gangliosidose GM1/patologia , Mitocôndrias/patologia , Adenina/análogos & derivados , Adenina/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Proteínas Reguladoras de Apoptose , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Autofagia/efeitos dos fármacos , Autofagia/genética , Proteína Beclina-1 , Encéfalo/ultraestrutura , Células Cultivadas , Modelos Animais de Doenças , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Inibidores Enzimáticos/farmacologia , Gangliosídeo G(M1)/metabolismo , Gangliosidose GM1/genética , Lisossomos/metabolismo , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/enzimologia , Paraquat/farmacologia , Proteínas Quinases/metabolismo , Proteínas/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR , beta-Galactosidase/deficiência
15.
Ann Neurol ; 62(6): 671-5, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17994547

RESUMO

Certain low-molecular-weight substrate analogs act both as in vitro competitive inhibitors of lysosomal hydrolases and as intracellular enhancers (chemical chaperones) by stabilization of mutant proteins. In this study, we performed oral administration of a chaperone compound N-octyl-4-epi-beta-valienamine to G(M1)-gangliosidosis model mice expressing R201C mutant human beta-galactosidase. A newly developed neurological scoring system was used for clinical assessment. N-Octyl-4-epi-beta-valienamine was delivered rapidly to the brain, increased beta-galactosidase activity, decreased ganglioside G(M1), and prevented neurological deterioration within a few months. No adverse effect was observed during this experiment. N-Octyl-4-epi-beta-valienamine will be useful for chemical chaperone therapy of human G(M1)-gangliosidosis.


Assuntos
Gangliosidose GM1/tratamento farmacológico , Gangliosidose GM1/fisiopatologia , Hexosaminas/uso terapêutico , Chaperonas Moleculares/uso terapêutico , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/fisiopatologia , Animais , Encéfalo/metabolismo , Gangliosidose GM1/metabolismo , Hexosaminas/farmacocinética , Humanos , Imuno-Histoquímica , Rim/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Chaperonas Moleculares/farmacocinética , Mutação , Sistema Nervoso/metabolismo , Concentração Osmolar , Distribuição Tecidual , beta-Galactosidase/deficiência , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
16.
Exp Anim ; 57(4): 385-95, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18633161

RESUMO

We assessed the possibility of C57BL/6-Tg (Meg1/Grb10)isn(Meg1 Tg) mice as a non-obese type 2 diabetes (2DM) animal model. Meg1 Tg mice were born normal, but their weight did not increase as much as normal after weaning and showed about 85% of normal size at 20 weeks of age. Body mass index of Meg1 Tg mice was also smaller than that of control mice. The glucose tolerance test and insulin tolerance test showed that Meg1 Tg mice had reduced ability to normalize the blood glucose level. Blood urea nitrogen (BUN) in Meg1 Tg mice (19.6 +/- 1.2 mg/dl) was significantly lower than in controls (22.0 +/- 0.8 mg/dl), while plasma triglyceride, insulin, adiponectin, and resistin levels were significantly higher (202.0 +/- 23.4 mg/dl vs 146.3 +/- 23.4 mg/dl, 152.4 +/- 16.3 pg/ml vs 88.1 +/- 16.9 pg/ml, 74.4 +/- 10.9 microg/ml vs 48.3 +/- 7.0 microg/ml, and 4.0 +/- 0.2 ng/ml vs 3.6 +/- 0.2 ng/ml, respectively). Body, visceral fat weight and liver weights were significantly lower (19.6 +/- 0.4 g vs 24.3 +/- 0.3 g, 376.7 +/- 29.6 mg to 507.5 +/- 23.0 mg, and 906.0 +/- 41.8 mg to 1,001.0 +/- 15.1 mg, respectively). Thus, hyperinsulinemia observed in Meg1 Tg mice indicates that their insulin signaling pathway is somehow inhibited. With high fat diet, the diabetes onset rate of Meg1 Tg mice increased up to 60%. These results suggest that Meg1 Tg mice resemble human 2DM.


Assuntos
Diabetes Mellitus Tipo 2/veterinária , Modelos Animais de Doenças , Camundongos Transgênicos , Adiponectina/sangue , Animais , Nitrogênio da Ureia Sanguínea , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Gorduras na Dieta , Teste de Tolerância a Glucose , Insulina/sangue , Insulina/farmacologia , Lipase/sangue , Camundongos , Resistina/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Yi Chuan ; 30(7): 877-84, 2008 Jul.
Artigo em Zh | MEDLINE | ID: mdl-18779131

RESUMO

Genetic diversity of Z:ZCLA Mongolian gerbils, wild Mongolian gerbils and 3 inbred M. gerbil strains was evaluated with 17 microsatellite loci. The genetic variabilities within and between populations were estimated. The results showed that 9 microsatellite DNA, AF200940, AF200941, AF200942, AF200945, AF200946, AF200947, D11Mit128, PKC, and SCN, were amplified efficiently both in Z:ZCLA M. gerbils and the wild M. gerbils. Forty-one alleles were amplified with the number of alleles per locus ranging from 1 to 7. The average expected heterozygosity (He) and polymorphism information content (PIC) of all the loci were 0.5032 and 0.4656, respectively. The mean effective allele number of Z:ZCLA M. gerbils and wild M. gerbils were 2.78 and 2.89. The PIC of Z:ZCLA M. gerbils and the wild M. gerbils were 0.3704 and 0.3893. In the 3 inbred M. gerbils strains, 8 microsatellite DNA were amplified efficiently with 11 alleles. It displayed heterozygosity in AF200941, AF200945, AF200946, D11Mit128, and SCN loci with fragment lengths from 140 to 215 bp; and homozygosity in AF200942, AF200946, and AF200947 with fragment lengths from 203 to 241 bp. All of the 8 microsatellite loci were monomorphic both within and among the strains. These results suggested that the moderate genetic diversity of the conventional closed colony of Z:ZCLA M. gerbils was observed; and inbred M. gerbils strains basically met the re-quest. Microsatellite markers can be used in monitoring of M. gerbils populations.


Assuntos
Variação Genética/genética , Gerbillinae/genética , Repetições de Microssatélites/genética , Animais , Reação em Cadeia da Polimerase
18.
Brain Dev ; 29(4): 210-6, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17027211

RESUMO

A large number of genetic disease model mice have been produced by genetic engineering. However, phenotypic analysis is not sufficient, particularly for brain dysfunction in neurogenetic diseases. We tried to develop a new assessment system mainly for motor and reflex functions in G(M1)-gangliosidosis model mice. Two genetically engineered model mouse strains were used for this study: the beta-galactosidase-deficient knockout mouse representing infantile G(M1)-gangliosidosis (severe form), and transgenic mouse representing juvenile G(M1)-gangliosidosis (mild form). We modified human child neurology techniques, and selected eleven tests for motor assessment and reflex testing. The test results were scored in four grades: 0 (normal), 1 (slightly abnormal), 2 (moderately abnormal), and 3 (severely abnormal). Both disease model mouse strains showed high scores even at the apparently pre-symptomatic stage of the disease, particularly with abnormal tail and hind limb postures. Individual and total test scores were well correlated with the progression of the disease. This method is simple, quick, and reproducible. The testing is sensitive enough to detect early neurological abnormalities, and will be useful for monitoring the natural clinical course and effect of therapeutic experiments in various neurogenetic disease model mice, such as chemical chaperone therapy for G(M1)-gangliosidosis model mice.


Assuntos
Gangliosidose GM1/fisiopatologia , Atividade Motora/genética , Mutação/fisiologia , Reflexo/fisiologia , Fatores Etários , Animais , Aprendizagem da Esquiva , Modelos Animais de Doenças , Feminino , Marcha , Gangliosidose GM1/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Exame Neurológico/métodos , Postura/fisiologia , Reflexo/genética , Fatores Sexuais , beta-Galactosidase/deficiência
19.
Elife ; 62017 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-29227248

RESUMO

Salt-inducible kinase 3 (SIK3) plays a crucial role in various aspects of metabolism. In the course of investigating metabolic defects in Sik3-deficient mice (Sik3-/-), we observed that circadian rhythmicity of the metabolisms was phase-delayed. Sik3-/- mice also exhibited other circadian abnormalities, including lengthening of the period, impaired entrainment to the light-dark cycle, phase variation in locomotor activities, and aberrant physiological rhythms. Ex vivo suprachiasmatic nucleus slices from Sik3-/- mice exhibited destabilized and desynchronized molecular rhythms among individual neurons. In cultured cells, Sik3-knockdown resulted in abnormal bioluminescence rhythms. Expression levels of PER2, a clock protein, were elevated in Sik3-knockdown cells but down-regulated in Sik3-overexpressing cells, which could be attributed to a phosphorylation-dependent decrease in PER2 protein stability. This was further confirmed by PER2 accumulation in the Sik3-/- fibroblasts and liver. Collectively, SIK3 plays key roles in circadian rhythms by facilitating phosphorylation-dependent PER2 destabilization, either directly or indirectly.


Assuntos
Relógios Circadianos , Regulação da Expressão Gênica , Proteínas Circadianas Period/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Comportamento Animal , Linhagem Celular , Humanos , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Serina-Treonina Quinases/deficiência
20.
Exp Anim ; 55(1): 65-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16508214

RESUMO

A transgene mapping technique (Noguchi et al., Exp. Anim. 53:103-111, 2004) is described that can be used to analyze transgene integration patterns in transgenic mice. The technique was used to reveal that a transgenic mouse line (GM1-sy#116) harbored inverted and direct tandem repeats of both intact and partial pCAGGS-based transgenes in the G2 region of chromosome 1. This complicated concatenation of transgenes may have been caused by simple end-joining of DNA constructs fragmented by exposure to UV transillumination during gel-purification, and by nuclease digestion inside zygote pronuclei. The results suggest that care should be taken to avoid unwanted fragmentation during the preparation of vector constructs.


Assuntos
Passeio de Cromossomo , Camundongos Transgênicos/genética , Transgenes/genética , Animais , Biblioteca Genômica , Genótipo , Camundongos , Recombinação Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA