Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pharmacol Rep ; 70(5): 1040-1046, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30144665

RESUMO

BACKGROUND: Aging is one of the most important inevitable risk factors of Alzheimer disease (AD). Oxidative stress plays a critical role in the process of aging. Curcumin has been proposed to improve neural damage, especially neurodegenerative injury, through its antioxidant and anti-inflammatory properties. Therefore, we investigated the effects of curcumin on acrolein-induced AD-like pathologies in HT22 cells. METHODS: HT22 murine hippocampal neuronal cells were treated with 25µM acrolein for 24h with or without pre-treating with curcumin at the selected optimum concentration (5µg/mL) for 30min. Cell viability and apoptosis were measured by CCK8 assay and flow cytometric analysis. Levels of glutathione (GSH), superoxide dismutase (SOD), and malondialdehyde (MDA) were detected by a GSH assay kit or commercial assay kits, respectively. Alterations in the expression of BDNF/TrkB and key enzymes involved in amyloid precursor protein (APP) metabolism were assessed by western blotting. RESULTS: Data showed that curcumin significantly reversed acrolein-induced oxidative stress indicated by depletion of GSH and SOD, and elevation of MDA. The findings also suggested curcumin's potential in protecting HT22 cells against acrolein through regulating the BDNF/TrkB signaling. In addition, acrolein-induced reduction in A-disintegrin and metalloprotease, and the increase of amyloid precursor protein, ß-secretase, and receptor for advanced glycation end products were reversed either, and most of them were nearly restored to the control levels by curcumin. CONCLUSION: These findings demonstrate the protective effects of curcumin on acrolein-induced neurotoxicity in vitro, which further suggests its potential role in the treatment of AD.


Assuntos
Acroleína/antagonistas & inibidores , Acroleína/toxicidade , Curcumina/farmacologia , Hipocampo/citologia , Fármacos Neuroprotetores/farmacologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/biossíntese , Animais , Apoptose/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Desintegrinas/metabolismo , Glutationa/metabolismo , Malondialdeído/metabolismo , Glicoproteínas de Membrana/metabolismo , Metaloproteases/metabolismo , Camundongos , Proteínas Tirosina Quinases/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Superóxido Dismutase/metabolismo
2.
J Biomed Res ; 32(1): 58-67, 2018 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-29353819

RESUMO

Meiosis-arrest female 1 (MARF1) is a recently identified key oogenic regulator essential for the maintenance of female fertility and genome integrity in mice. However, the detailed functions and the underlying mechanisms of MARF1 remain elusive. Here, in an attempt to create a mouse model expressing fluorescent protein-tagged MARF1 to facilitate further exploration of the roles of MARF1 in oocytes, we produced a Marf1-eGFP knockin (KI) mouse line in which the C-terminal structure and function of MARF1 were interfered by its fusing eGFP peptide. Using these Marf1-eGFP-KI mice, we revealed, unexpectedly, the functions of MARF1 in the control of oocyte meiotic division. We found that the Marf1-eGFP-KI females ovulated mature oocytes with severe meiotic and developmental defects, and thus were infertile. Moreover, meiotic reinitiation was delayed while meiotic completion was accelerated in the KI-oocytes, which was coincident with the increased incidence of oocyte aneuploidy. Therefore, MARF1 is indispensable for maintaining the fidelity of homolog segregation during oocyte maturation, and this function relies on its C-terminal domains.

3.
Neurotoxicology ; 65: 44-51, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29355571

RESUMO

OBJECTIVE: Acrolein, a highly reactive unsaturated aldehyde, is a ubiquitous environmental pollutant and oxidative damage induced by acrolein is hypothesized to involve in the etiology of Alzheimer's disease (AD). Calorie restriction (CR) is the only non-genetic intervention that has consistently been verified to retard aging by ameliorating oxidative stress. Therefore, we investigated the effects of CR on acrolein-induced neurotoxicity in Sprague-Dawley (SD) rats. METHODS: A total of 45 weaned and specific-pathogen-free SD rats (male, weighing 180-220 g) were gavage-fed with acrolein (2.5 mg/kg/day) and fed ab libitum of 10 g/day or 7 g/day (representing 30% CR regimen), or gavage-fed with same volume of tap water and fed al libitum as vehicle control for 12 weeks. After behavioral test conducted by Morris Water Maze, SD rats were sacrificed and brain tissues were prepared for histochemical evaluation and Western blotting to detect alterations in oxidative stress, BDNF/TrkB pathway and key enzymes involved in amyloid precursor protein (APP) metabolism. RESULTS: Treatment with 30% CR in SD rats significantly attenuated acrolein-induced cognitive impairment. Oxidative damage including deletion of glutathione and superoxide dismutase and sharp rise in malondialdehyde were notably improved by 30% CR. Further study suggested that 30% CR showed protective effects against acrolein by modulating BDNF/TrkB signaling pathways. Moreover, 30% CR restored acrolein-induced changes of APP, ß-secretase, α-secretase and receptor for advanced glycation end products. CONCLUSION: These findings suggest that CR may provide a promising approach for the treatment of AD, targeting acrolein.


Assuntos
Acroleína/toxicidade , Restrição Calórica , Disfunção Cognitiva/prevenção & controle , Síndromes Neurotóxicas/prevenção & controle , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Córtex Cerebral/metabolismo , Disfunção Cognitiva/induzido quimicamente , Glutationa/metabolismo , Hipocampo/metabolismo , Masculino , Malondialdeído/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Receptor trkB/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo
4.
PLoS One ; 12(8): e0183837, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28859131

RESUMO

Mouse models provide a valuable tool for exploring pathogenic mechanisms underlying inherited human disease. Here, we describe seven mouse models identified through the Translational Vision Research Models (TVRM) program, each carrying a new allele of a gene previously linked to retinal developmental and/or degenerative disease. The mutations include four alleles of three genes linked to human nonsyndromic ocular diseases (Aipl1tvrm119, Aipl1tvrm127, Rpgrip1tvrm111, RhoTvrm334) and three alleles of genes associated with human syndromic diseases that exhibit ocular phentoypes (Alms1tvrm102, Clcn2nmf289, Fkrptvrm53). Phenotypic characterization of each model is provided in the context of existing literature, in some cases refining our current understanding of specific disease attributes. These murine models, on fixed genetic backgrounds, are available for distribution upon request and may be useful for understanding the function of the gene in the retina, the pathological mechanisms induced by its disruption, and for testing experimental approaches to treat the corresponding human ocular diseases.


Assuntos
Oftalmopatias/genética , Degeneração Retiniana/genética , Pesquisa Translacional Biomédica , Visão Ocular/genética , Alelos , Animais , Canais de Cloro CLC-2 , Proteínas de Ciclo Celular , Canais de Cloreto/genética , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Oftalmopatias/patologia , Humanos , Camundongos , Mutação , Pentosiltransferases , Proteínas/genética , Retina/patologia , Degeneração Retiniana/patologia , Transferases
5.
Am J Pathol ; 186(7): 1925-1938, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27207593

RESUMO

The nicotinamide nucleotide adenylyltransferase 1 (NMNAT1) enzyme is essential for regenerating the nuclear pool of NAD(+) in all nucleated cells in the body, and mounting evidence also suggests that it has a separate role in neuroprotection. Recently, mutations in the NMNAT1 gene were associated with Leber congenital amaurosis, a severe retinal degenerative disease that causes blindness during infancy. Availability of a reliable mammalian model of NMNAT1-Leber congenital amaurosis would assist in determining the mechanisms through which disruptions in NMNAT1 lead to retinal cell degeneration and would provide a resource for testing treatment options. To this end, we identified two separate N-ethyl-N-nitrosourea-generated mouse lines that harbor either a p.V9M or a p.D243G mutation. Both mouse models recapitulate key aspects of the human disease and confirm the pathogenicity of mutant NMNAT1. Homozygous Nmnat1 mutant mice develop a rapidly progressing chorioretinal disease that begins with photoreceptor degeneration and includes attenuation of the retinal vasculature, optic atrophy, and retinal pigment epithelium loss. Retinal function deteriorates in both mouse lines, and, in the more rapidly progressing homozygous Nmnat1(V9M) mutant mice, the electroretinogram becomes undetectable and the pupillary light response weakens. These mouse models offer an opportunity for investigating the cellular mechanisms underlying disease pathogenesis, evaluating potential therapies for NMNAT1-Leber congenital amaurosis, and conducting in situ studies on NMNAT1 function and NAD(+) metabolism.


Assuntos
Modelos Animais de Doenças , Amaurose Congênita de Leber/genética , Amaurose Congênita de Leber/fisiopatologia , Nicotinamida-Nucleotídeo Adenililtransferase/genética , Animais , Genótipo , Humanos , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase
6.
Adv Exp Med Biol ; 854: 177-83, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26427409

RESUMO

Mouse models provide important resources for many areas of vision research, pertaining to retinal development, retinal function and retinal disease. The Translational Vision Research Models (TVRM) program uses chemical mutagenesis to generate new mouse models for vision research. In this chapter, we report the identification of mouse models for Grm1, Grk1 and Lrit3. Each of these is characterized by a primary defect in the electroretinogram. All are available without restriction to the research community.


Assuntos
Predisposição Genética para Doença/genética , Mutação , Retina/metabolismo , Doenças Retinianas/genética , Alelos , Animais , Modelos Animais de Doenças , Eletrorretinografia , Oftalmopatias/diagnóstico , Oftalmopatias/genética , Oftalmopatias/fisiopatologia , Receptor Quinase 1 Acoplada a Proteína G/genética , Testes Genéticos/métodos , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Mutagênese , Receptores de Glutamato Metabotrópico/genética , Retina/patologia , Retina/fisiopatologia , Doenças Retinianas/diagnóstico , Pesquisa Translacional Biomédica/métodos , Visão Ocular/genética , Visão Ocular/fisiologia
7.
Proc Natl Acad Sci U S A ; 112(42): 12962-7, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26438849

RESUMO

Sphingolipids typically have an 18-carbon (C18) sphingoid long chain base (LCB) backbone. Although sphingolipids with LCBs of other chain lengths have been identified, the functional significance of these low-abundance sphingolipids is unknown. The LCB chain length is determined by serine palmitoyltransferase (SPT) isoenzymes, which are trimeric proteins composed of two large subunits (SPTLC1 and SPTLC2 or SPTLC3) and a small subunit (SPTssa or SPTssb). Here we report the identification of an Sptssb mutation, Stellar (Stl), which increased the SPT affinity toward the C18 fatty acyl-CoA substrate by twofold and significantly elevated 20-carbon (C20) LCB production in the mutant mouse brain and eye, resulting in surprising neurodegenerative effects including aberrant membrane structures, accumulation of ubiquitinated proteins on membranes, and axon degeneration. Our work demonstrates that SPT small subunits play a major role in controlling SPT activity and substrate affinity, and in specifying sphingolipid LCB chain length in vivo. Moreover, our studies also suggest that excessive C20 LCBs or C20 LCB-containing sphingolipids impair protein homeostasis and neural functions.


Assuntos
Carbono/química , Mutação , Doenças Neurodegenerativas/enzimologia , Serina C-Palmitoiltransferase/química , Sequência de Aminoácidos , Animais , Humanos , Camundongos , Dados de Sequência Molecular , Doenças Neurodegenerativas/genética , Homologia de Sequência de Aminoácidos , Serina C-Palmitoiltransferase/genética , Ubiquitinação
8.
Curr Med Res Opin ; 31(12): 2313-23, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26402735

RESUMO

OBJECTIVE: New P2Y12 inhibitors, classified as oral (prasugrel and ticagrelor) and intravenous (cangrelor and elinogrel) drugs, have shown improved antithrombotic effects compared with clopidogrel in patients with acute coronary syndrome (ACS) or patients undergoing percutaneous coronary intervention (PCI) in landmark trials. The purpose of this study was to perform a meta-analysis of randomized trials that compared new P2Y12 inhibitors with clopidogrel to determine their efficacy and safety in patients undergoing PCI. METHODS: Randomized controlled trials of at least 4 weeks, comparing new P2Y12 inhibitors with clopidogrel in PCI, were identified using the electronic databases Cochrane Central Register of Controlled Trials, Medline, PubMed, Web of Science, and Google Scholar from January 1, 1980, to July 31, 2014. MAIN OUTCOME MEASURES: The primary efficacy endpoints were all-cause death and major adverse cardiovascular events (MACEs). The primary safety endpoint was thrombolysis in myocardial infarction (TIMI) major bleeding. RESULTS: Twelve studies including 71,097 patients met the inclusion criteria. New P2Y12 inhibitors significantly reduced all-cause death (odds ratio [OR]: 0.81; 95% confidence interval [CI] 0.73-0.90, p < 0.0001), MACEs (OR 0.81; 95% CI 0.73-0.90, p < 0.0001), stent thrombosis (OR 0.58; 95% CI 0.49-0.69, p < 0.00001), myocardial infarctions (OR 0.87; 95% CI 0.76-0.99, p = 0.03) and cardiovascular death (OR 0.82; 95% CI 0.73-0.92, p = 0.001) compared with clopidogrel. There were no significant differences between stroke (OR 0.87; 95% CI 0.72-1.05, p = 0.14) and major bleeding events (OR 1.22; 95% CI 0.99-1.52, p = 0.06) between the new P2Y12 inhibitor and clopidogrel groups. CONCLUSION: New P2Y12 inhibitors decreased death in patients undergoing PCI compared with clopidogrel with a considerable safety and tolerability profile; however, the risk/benefit ratio of ischemic and bleeding events should be further investigated.


Assuntos
Inibidores da Agregação Plaquetária/uso terapêutico , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Ticlopidina/análogos & derivados , Síndrome Coronariana Aguda/terapia , Clopidogrel , Hemorragia/induzido quimicamente , Humanos , Infarto do Miocárdio/terapia , Intervenção Coronária Percutânea/métodos , Inibidores da Agregação Plaquetária/efeitos adversos , Antagonistas do Receptor Purinérgico P2Y/efeitos adversos , Ensaios Clínicos Controlados Aleatórios como Assunto , Acidente Vascular Cerebral/epidemiologia , Acidente Vascular Cerebral/etiologia , Ticlopidina/efeitos adversos , Ticlopidina/uso terapêutico
9.
Nutr J ; 13: 110, 2014 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-25424121

RESUMO

BACKGROUND: The present study aimed to determine that whether L-carnitine infusion could ameliorate fasting-induced adverse effects and improve outcomes. METHOD: In this 7-day, randomized, single-blind, placebo-controlled, pilot study, 15 metabolic syndrome (MetS) patients (11/4 F/M; age 46.9 ± 9.14 years; body mass index [BMI] 28.2 ± 1.8 kg/m2) were in the L-carnitine group (LC) and 15 (10/5 F/M; age 46.8 ± 10.9 years; BMI 27.1 ± 2.3 kg/m2) were in the control group (CT). All participants underwent a 5-day modified fasting therapy introduced with 2-day moderate calorie restriction. Patients in the LC group received 4 g/day of intravenous L-carnitine, while patients in the CT group were injected with saline. Blood pressure (BP), anthropometric characteristics, markers of liver function, metabolic indices (plasma glucose, lipid profiles, uric acid, free fatty acid and insulin) and hypersensitivity C-reactive protein were measured. Perceived hunger was recorded daily by self-rating visual analogue scales. Fatigue was evaluated by Wessely and Powell scores. RESULTS: In contrast to the CT group, total cholesterol, alanine aminotransferase, systolic and diastolic BP did not change significantly in the LC group after prolonged fasting. There were significant differences in weight loss (LC -4.6 ± 0.9 vs. CT -3.2 ± 1.1 kg, P = 0.03), and waist circumference (LC -5.0 ± 2.2 vs. CT -1.7 ± 1.16 cm, P < 0.001), waist hip ratio (LC -0.023 ± 0.017 vs. CT 0.012 ± 0.01, P < 0.001), insulin concentration (LC -9.9 ± 3.58 vs. CT -6.32 ± 3.44 µU/mL, P = 0.046), and γ-glutamyltransferase concentration (LC -7.07 ± 6.82 vs. CT -2.07 ± 4.18, P = 0.024). Perceived hunger scores were significantly increased (P < 0.05) in the CT group during starvation, which was alleviated with L-carnitine administration in the LC group. Physical fatigue (LC -3.2 ± 3.17 vs. CT 1.8 ± 2.04, P < 0.001) and fatigue severity (LC -11.6 ± 8.38 vs. CT 8.18 ± 7.32, P < 0.001) were significantly reduced in the LC group but were aggravated in the CT group. CONCLUSION: Intravenous L-carnitine can ameliorate fasting-induced hunger, fatigue, cholesterol abnormalities and hepatic metabolic changes and facilitate fasting-induced weight loss in MetS patients. TRIAL REGISTRATION: ChiCTR-TNRC-12002835.


Assuntos
Carnitina/administração & dosagem , Jejum/efeitos adversos , Fadiga/tratamento farmacológico , Fome/efeitos dos fármacos , Síndrome Metabólica/metabolismo , Administração Intravenosa , Adulto , Alanina Transaminase/sangue , Glicemia/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Índice de Massa Corporal , Proteína C-Reativa/metabolismo , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Dieta , Exercício Físico , Ácidos Graxos não Esterificados/sangue , Feminino , Humanos , Insulina/sangue , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Método Simples-Cego , Resultado do Tratamento , Ácido Úrico/sangue , Circunferência da Cintura , Redução de Peso/efeitos dos fármacos , gama-Glutamiltransferase/sangue
10.
Am J Pathol ; 184(10): 2721-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25088982

RESUMO

Mutations in the RP1 gene can cause retinitis pigmentosa. We identified a spontaneous L66P mutation caused by two adjacent point mutations in the Rp1 gene in a colony of C57BL/6J mice. Mice homozygous for the L66P mutation exhibited slow, progressive photoreceptor degeneration throughout their lifespan. Optical coherence tomography imaging found abnormal photoreceptor reflectivity at 1 month of age. Histology found shortening and disorganization of the photoreceptor inner and outer segments and progressive thinning of the outer nuclear layer. Electroretinogram a- and b-wave amplitudes were decreased with age. Western blot analysis found that the quantity and size of the mutated retinitis pigmentosa 1 (RP1) protein were normal. However, immunohistochemistry found that the mutant Rp1 protein partially mislocalized to the transition zone of the shortened axonemes. This mutation disrupted colocalization with cytoplasmic microtubules in vitro. In conclusion, the L66P mutation in the first doublecortin domain of the Rp1 gene impairs Rp1 protein localization and function, leading to abnormalities in photoreceptor outer segment structure and progressive photoreceptor degeneration. This is the first missense mutation in Rp1 shown to cause retinal degeneration. It provides a unique, slowly progressive photoreceptor degeneration model that mirrors the slow degeneration kinetics in most patients with retinitis pigmentosa.


Assuntos
Axonema/metabolismo , Proteínas do Olho/genética , Proteínas Associadas aos Microtúbulos/genética , Degeneração Retiniana/genética , Animais , Células COS , Chlorocebus aethiops , Eletrorretinografia , Proteínas do Olho/metabolismo , Feminino , Homozigoto , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação de Sentido Incorreto , Células Fotorreceptoras/metabolismo , Degeneração Retiniana/metabolismo
11.
12.
J Ophthalmol ; 2011: 391384, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21052544

RESUMO

The need for mouse models, with their well-developed genetics and similarity to human physiology and anatomy, is clear and their central role in furthering our understanding of human disease is readily apparent in the literature. Mice carrying mutations that alter developmental pathways or cellular function provide model systems for analyzing defects in comparable human disorders and for testing therapeutic strategies. Mutant mice also provide reproducible, experimental systems for elucidating pathways of normal development and function. Two programs, the Eye Mutant Resource and the Translational Vision Research Models, focused on providing such models to the vision research community are described herein. Over 100 mutant lines from the Eye Mutant Resource and 60 mutant lines from the Translational Vision Research Models have been developed. The ocular diseases of the mutant lines include a wide range of phenotypes, including cataracts, retinal dysplasia and degeneration, and abnormal blood vessel formation. The mutations in disease genes have been mapped and in some cases identified by direct sequencing. Here, we report 3 novel alleles of Crx(tvrm65), Rp1(tvrm64), and Rpe65(tvrm148) as successful examples of the TVRM program, that closely resemble previously reported knockout models.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...