Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Appl Opt ; 61(34): 10228, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36606785

RESUMO

This publisher's note serves to correct an error in Appl. Opt. 58, 3495 (2019)APOPAI0003-693510.1364/AO.58.003495.

2.
Biochim Biophys Acta Mol Basis Dis ; 1865(11): 165531, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31398466

RESUMO

Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder worldwide. While the causes of AD are unclear, several risk factors have been identified, including impaired glycemic control, which significantly increases the risk of cognitive decline and AD. In vitro and in vivo studies show that human adenovirus 36 (Ad36) improves glycemic control by increasing cellular glucose uptake in cells, experimental animal models and in humans who are naturally exposed to the virus. This study, tested improvement in glycemic control by Ad36 and delay in onset of cognitive decline in APPswe transgenic mice (Tg2576 line), a model of genetic predisposition to impaired glycemic control and AD. Three-month old APPswe mice were divided into Ad36 infected (Ad36) or mock infected (control) groups and baseline glycemic control measured by glucose tolerance test (GTT) prior to infection. Changes in glycemic control were determined 10- and 24-week post infection. Serum insulin was also measured during GTT. Cognition was determined by Y-maze test, while motor coordination and skill acquisition by rotarod test. Glycemic control as determined by GTT showed less deterioration in Ad36 infected mice over time, accompanied by a significant attenuation of cognitive decline. Analysis of brain tissue lysate showed significantly reduced levels of amyloid beta 42 in Ad36 mice relative to control mice. Golgi-Cox staining analysis also revealed reduced dendritic spines and synaptic gene expression in control mice compared to Ad36 infected mice. This proof of concept study shows that in a mouse model of AD, Ad36 improves glycemic control and ameliorates cognitive decline.


Assuntos
Infecções por Adenoviridae/complicações , Adenoviridae/fisiologia , Doença de Alzheimer/complicações , Disfunção Cognitiva/complicações , Infecções por Adenoviridae/sangue , Infecções por Adenoviridae/patologia , Doença de Alzheimer/sangue , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/análise , Animais , Glicemia/análise , Disfunção Cognitiva/sangue , Disfunção Cognitiva/patologia , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Teste de Tolerância a Glucose , Humanos , Camundongos Transgênicos , Fatores de Proteção
3.
Appl Opt ; 58(13): 3495-3500, 2019 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-31044848

RESUMO

A passively Q-switched ytterbium-doped fiber laser (YDFL) operating at 1062 nm was demonstrated by using a segment of 20 cm titanium dioxide-doped fiber saturable absorber (TiO2DF SA). The Q-switched YDFL emerged stably with tunable repetition rates ranging from 32 kHz to 53 kHz as the pump power rose from 109 mW to 233 mW. Within this range of pump power, a maximum output power of 10.1 mW, maximum peak power of 75 mW, and maximum pulse energy of 191 nJ were obtained. The narrowest pulse width of 2.55 µs was attained at the maximum pump power of 233 mW, while the signal-to-noise ratio of the fundamental frequency was 47 dB. This demonstration reveals that the proposed TiO2DF SA is feasible for constructing a flexible and reliably stable Q-switched pulsed fiber laser in the 1-micrometer region.

4.
Prog Mol Biol Transl Sci ; 146: 127-171, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28253983

RESUMO

Aging is a normal process of living being. It has been reported that multiple cellular changes, including oxidative damage/mitochondrial dysfunction, telomere shortening, inflammation, may accelerate the aging process, leading to cellular senescence. These cellular changes induce age-related human diseases, including Alzheimer's, Parkinson's, multiple sclerosis, amyotrophic lateral sclerosis, cardiovascular, cancer, and skin diseases. Changes in somatic and germ-line DNA and epigenetics are reported to play large roles in accelerating the onset of human diseases. Cellular mechanisms of aging and age-related diseases are not completely understood. However, recent discoveries in molecular biology have revealed that microRNAs (miRNAs) are potential indicators of aging, cellular senescence, and Alzheimer's disease (AD). The purpose of our chapter is to highlight recent advancements in miRNAs and their involvement in cellular changes in aging, cellular senescence, and AD. This chapter also critically evaluates miRNA-based therapeutic drug targets for aging and age-related diseases, particularly Alzheimer's.


Assuntos
Envelhecimento/genética , Doença de Alzheimer/genética , Senescência Celular/genética , MicroRNAs/metabolismo , Animais , Humanos , Degeneração Neural/genética , Degeneração Neural/patologia , Transdução de Sinais/genética
5.
Prog Mol Biol Transl Sci ; 146: 13-46, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28253984

RESUMO

Mitochondria are complex, intercellular organelles present in the cells and are involved in multiple roles including ATP formation, free radicals generation and scavenging, calcium homeostasis, cellular differentiation, and cell death. Many studies depicted the involvement of mitochondrial dysfunction and oxidative damage in aging and pathogenesis of age-related metabolic disorders and neurodegenerative diseases. Remarkable advancements have been made in understanding the structure, function, and physiology of mitochondria in metabolic disorders such as diabetes, obesity, cardiovascular diseases, and stroke. Further, much progress has been done in the improvement of therapeutic strategies, including lifestyle interventions, pharmacological, and mitochondria-targeted therapeutic approaches. These strategies were mainly focused to reduce the mitochondrial dysfunction caused by oxidative stress and to retain the mitochondrial health in various diseases. In this chapter, we have highlighted the involvement of mitochondrial dysfunction in the pathophysiology of various disorders and recent progress in the development of mitochondria-targeted molecules as therapeutic measures for metabolic disorders.


Assuntos
Envelhecimento/patologia , Doenças Metabólicas/terapia , Mitocôndrias/patologia , Estresse Oxidativo , Animais , Humanos , Doenças Metabólicas/epidemiologia , Modelos Biológicos , Terapia de Alvo Molecular
6.
Prog Mol Biol Transl Sci ; 146: 173-201, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28253985

RESUMO

Alzheimer's disease (AD) is the most common multifactorial mental illness affecting the elderly population in the world. Its prevalence increases as person ages. There is no known drug or agent that can delay or prevent the AD and its progression. Extensive research has revealed that multiple cellular pathways involved, including amyloid beta production, mitochondrial structural and functional changes, hyperphosphorylation of Tau and NFT formation, inflammatory responses, and neuronal loss in AD pathogenesis. Amyloid beta-induced synaptic damage, mitochondrial abnormalities, and phosphorylated Tau are major areas of present research investigations. Synaptic pathology and mitochondrial oxidative damage are early events in disease process. In this chapter, a systematic literature survey has been conducted and presented a summary of antioxidants used in (1) AD mouse models, (2) elderly populations, and (3) randomized clinical trials in AD patients. This chapter highlights the recent progress in developing and testing mitochondria-targeted molecules using AD cell cultures and AD mouse models. This chapter also discusses recent research on AD pathogenesis and therapeutics, focusing on mitochondria-targeted molecules as potential therapeutic targets to delay or prevent AD progression.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Mitocôndrias/metabolismo , Terapia de Alvo Molecular , Animais , Ensaios Clínicos como Assunto , Humanos , Neurônios/metabolismo , Sinapses/patologia
7.
Prog Mol Biol Transl Sci ; 146: 47-94, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28253991

RESUMO

MicroRNAs (miRNAs) are found in the circulatory biofluids considering the important molecules for biomarker study in aging and age-related diseases. Blood or blood components (serum/plasma) are primary sources of circulatory miRNAs and can release these in cell-free form either bound with some protein components or encapsulated with microvesicle particles, called exosomes. miRNAs are quite stable in the peripheral circulation and can be detected by high-throughput techniques like qRT-PCR, microarray, and sequencing. Intracellular miRNAs could modulate mRNA activity through target-specific binding and play a crucial role in intercellular communications. At a pathological level, changes in cellular homeostasis lead to the modulation of molecular function of cells; as a result, miRNA expression is deregulated. Deregulated miRNAs came out from cells and frequently circulate in extracellular body fluids as part of various human diseases. Most common aging-associated diseases are cardiovascular disease, cancer, arthritis, dementia, cataract, osteoporosis, diabetes, hypertension, and neurodegenerative diseases such as Alzheimer's disease, Huntington's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Variation in the miRNA signature in a diseased peripheral circulatory system opens up a new avenue in the field of biomarker discovery. Here, we measure the biomarker potential of circulatory miRNAs in aging and various aging-related pathologies. However, further more confirmatory researches are needed to elaborate these findings at the translation level.


Assuntos
Envelhecimento/genética , Doença , MicroRNAs/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Humanos , MicroRNAs/sangue , Modelos Biológicos
8.
Prog Mol Biol Transl Sci ; 146: 95-126, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28253992

RESUMO

Stroke is a very common neurological disease, and it occurs when the blood supply to part of the brain is interrupted and the subsequent shortage of oxygen and nutrients causes damage to the brain tissue. Stroke is the second leading cause of death and the third leading cause of disability-adjusted life years. The occurrence of stroke increases with age, but anyone at any age can suffer a stroke. Stroke can be broadly classified in two major clinical types: ischemic stroke (IS) and hemorrhagic stroke. Research also revealed that stroke, vascular dementia (VaD), and Alzheimer's disease (AD) increase with a number of modifiable factors, and most strokes can be prevented and/or controlled through pharmacological or surgical interventions and lifestyle changes. The pathophysiology of stroke, VaD, and AD is complex, and recent molecular and postmortem brain studies have revealed that multiple cellular changes have been implicated, including inflammatory responses, microRNA alterations, and marked changes in brain proteins. These molecular and cellular changes provide new information for developing therapeutic strategies for stroke and related vascular disorders treatment. IS is the major risk factor for VaD and AD. This chapter summarizes the (1) links among stroke-VaD-AD; (2) updates the latest developments of research in identifying protein biomarkers in peripheral and central nervous system tissues; and (3) critically evaluates miRNA profile and function in human blood samples, animal, and postmortem brains.


Assuntos
Doença de Alzheimer/genética , Biomarcadores/metabolismo , Demência Vascular/genética , Acidente Vascular Cerebral/genética , Animais , Humanos , Fatores de Risco , Transdução de Sinais/genética
9.
Curr Alzheimer Res ; 8(4): 393-409, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21470101

RESUMO

Mitochondria are cytoplasmic organelles responsible for life and death. Extensive evidence from animal models, postmortem brain studies of and clinical studies of aging and neurodegenerative diseases suggests that mitochondrial function is defective in aging and neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Several lines of research suggest that mitochondrial abnormalities, including defects in oxidative phosphorylation, increased accumulation of mitochondrial DNA defects, impaired calcium influx, accumulation of mutant proteins in mitochondria, and mitochondrial membrane potential dissipation are important cellular changes in both early and late-onset neurodegenerative diseases. Further, emerging evidence suggests that structural changes in mitochondria, including increased mitochondrial fragmentation and decreased mitochondrial fusion, are critical factors associated with mitochondrial dysfunction and cell death in aging and neurodegenerative diseases. This paper discusses research that elucidates features of mitochondria that are associated with cellular dysfunction in aging and neurodegenerative diseases and discusses mitochondrial structural and functional changes, and abnormal mitochondrial dynamics in neurodegenerative diseases. It also outlines mitochondria-targeted therapeutics in neurodegenerative diseases.


Assuntos
Encéfalo/ultraestrutura , DNA Mitocondrial/fisiologia , Mitocôndrias/patologia , Doenças Mitocondriais/etiologia , Doenças Neurodegenerativas/complicações , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Humanos , Modelos Biológicos , Doenças Neurodegenerativas/patologia
10.
Mol Cell Neurosci ; 22(3): 285-97, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12691731

RESUMO

Huntington's disease is due to an expansion of CAG repeats in the huntingtin gene. Huntingtin interacts with several proteins including glyceraldehyde-3-phosphate dehydrogenase (GAPDH). We performed immunohistochemical analysis of GAPDH expression in the brains of transgenic mice carrying the huntingtin gene with 89 CAG repeats. In all wild-type animals examined, GAPDH was evenly distributed among the different cell types throughout the brain. In contrast, the majority of transgenic mice showed GAPDH overexpression, with the most prominent GAPDH changes observed in the caudate putamen, globus pallidus, neocortex, and hippocampal formation. Double staining for NeuN and GFAP revealed that GAPDH overexpression occurred exclusively in neurons. Nissl staining analysis of the neocortex and caudate putamen indicated 24 and 27% of cell loss in transgenic mice, respectively. Subcellular fluorescence analysis revealed a predominant increase in GAPDH immunostaining in the nucleus. Thus, we conclude that mutation of huntingtin is associated with GAPDH overexpression and nuclear translocation in discrete populations of brain neurons.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/genética , Doença de Huntington/genética , Doença de Huntington/metabolismo , Animais , Núcleo Caudado/enzimologia , Núcleo Caudado/patologia , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Proteína Huntingtina , Doença de Huntington/patologia , Camundongos , Camundongos Transgênicos , Neocórtex/enzimologia , Neocórtex/patologia , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Expansão das Repetições de Trinucleotídeos
11.
J Neurosci ; 21(23): 9112-23, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11717344

RESUMO

Neurons in Huntington's disease exhibit selective morphological and subcellular alterations in the striatum and cortex. The link between these neuronal changes and behavioral abnormalities is unclear. We investigated relationships between essential neuronal changes that predict motor impairment and possible involvement of the corticostriatal pathway in developing behavioral phenotypes. We therefore generated heterozygote mice expressing the N-terminal one-third of huntingtin with normal (CT18) or expanded (HD46, HD100) glutamine repeats. The HD mice exhibited motor deficits between 3 and 10 months. The age of onset depended on an expanded polyglutamine length; phenotype severity correlated with increasing age. Neuronal changes in the striatum (nuclear inclusions) preceded the onset of phenotype, whereas cortical changes, especially the accumulation of huntingtin in the nucleus and cytoplasm and the appearance of dysmorphic dendrites, predicted the onset and severity of behavioral deficits. Striatal neurons in the HD mice displayed altered responses to cortical stimulation and to activation by the excitotoxic agent NMDA. Application of NMDA increased intracellular Ca(2+) levels in HD100 neurons compared with wild-type neurons. Results suggest that motor deficits in Huntington's disease arise from cumulative morphological and physiological changes in neurons that impair corticostriatal circuitry.


Assuntos
Comportamento Animal , Córtex Cerebral/fisiopatologia , Corpo Estriado/fisiopatologia , Doença de Huntington/fisiopatologia , Neurônios/metabolismo , Idade de Início , Animais , Cálcio/metabolismo , Núcleo Celular/patologia , Córtex Cerebral/patologia , Corpo Caloso/fisiopatologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Dendritos/patologia , Modelos Animais de Doenças , Progressão da Doença , Eletrofisiologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Heterozigoto , Proteína Huntingtina , Doença de Huntington/patologia , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenótipo , Receptores de N-Metil-D-Aspartato/metabolismo , Expansão das Repetições de Trinucleotídeos
12.
Exp Neurol ; 169(2): 340-50, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11358447

RESUMO

Mitochondrial defects, which occur in the brain of late-stage Huntington's disease (HD) patients, have been proposed to underlie the selective neuronal loss in the disease. To shed light on the possible role of mitochondrial energy impairment in the early phases of HD pathophysiology, we carried out Golgi impregnation and quantitative histochemical/biochemical studies in HD full-length cDNA transgenic mice that were symptomatic but had not developed to a stage in which neuronal loss could be documented. Golgi staining showed morphologic abnormalities that included a significant decrease in the number of dendritic spines and a thickening of proximal dendrites in striatal and cortical neurons. In contrast, measurements of mitochondrial electron transport Complexes I-IV did not reveal changes in the striatum and cerebral cortex in these mice. Examination of the neostriatum and cerebral cortex in human presymptomatic and pathological Grade 1 HD cases also showed no change in the activity of mitochondrial Complexes I-IV. These data suggest that dendritic alterations precede irreversible cell loss in HD, and that mitochondrial energy impairment is a consequence, rather than a cause, of early neuropathological changes.


Assuntos
Corpo Estriado/patologia , Dendritos/patologia , Doença de Huntington/genética , Mitocôndrias/metabolismo , Degeneração Neural/genética , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Proteínas Nucleares/genética , Córtex Somatossensorial/metabolismo , Animais , Núcleo Caudado/patologia , Corantes , DNA Complementar , Dendritos/ultraestrutura , Metabolismo Energético , Lateralidade Funcional , Complexo de Golgi/patologia , Complexo de Golgi/ultraestrutura , Heterozigoto , Humanos , Proteína Huntingtina , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Mitocôndrias/patologia , Atividade Motora , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Putamen/patologia , Córtex Somatossensorial/patologia
13.
J Biol Chem ; 276(27): 24713-8, 2001 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-11319238

RESUMO

Increased glutamate-mediated excitotoxicity seems to play an important role in the pathogenesis of Huntington's disease (Tabrizi, S. J., Cleeter, M. W., Xuereb, J., Taaman, J. W., Cooper, J. M., and Schapira, A. H. (1999) Ann. Neurol. 45, 25-32). However, how polyglutamine expansion in huntingtin promotes glutamate-mediated excitotoxicity remains a mystery. In this study we provide evidence that (i) normal huntingtin is associated with N-methyl-d-aspartate (NMDA) and kainate receptors via postsynaptic density 95 (PSD-95), (ii) the SH3 domain of PSD-95 mediates its binding to huntingtin, and (iii) polyglutamine expansion interferes with the ability of huntingtin to interact with PSD-95. The expression of polyglutamine-expanded huntingtin causes sensitization of NMDA receptors and promotes neuronal apoptosis induced by glutamate. The addition of the NMDA receptor antagonist significantly attenuates neuronal toxicity induced by glutamate and polyglutamine-expanded huntingtin. The overexpression of normal huntingtin significantly inhibits neuronal toxicity mediated by NMDA or kainate receptors. Our results demonstrate that polyglutamine expansion impairs the ability of huntingtin to bind PSD-95 and inhibits glutamate-mediated excitotoxicity. These changes may be essential for the pathogenesis of Huntington's disease.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Linhagem Celular , Proteína 4 Homóloga a Disks-Large , Humanos , Proteína Huntingtina , Doença de Huntington/fisiopatologia , Marcação In Situ das Extremidades Cortadas , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Peso Molecular , Neurônios/efeitos dos fármacos , Ligação Proteica , Ratos , Receptores de Ácido Caínico/metabolismo , Transfecção , Domínios de Homologia de src
14.
Hum Biol ; 72(3): 499-510, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10885195

RESUMO

This paper presents data on the distribution of 3 amplified fragment length polymorphisms (D1S80, APOB, and YNZ22) in 5 populations of Central India. Using the polymerase chain reaction technique, 3 caste (Brahmin, Khatri, and Dhimer) and 2 tribal (Gond and Baiga) populations were studied for the 3 loci. The allelic variations observed in the caste populations are compatible with those of many Caucasian populations, but the caste populations showed significant overall and interpopulation variability within the region. D1S80 allele *24 varied from 32% (Dhimers) to 42% (Brahmins). Allele *18 was not observed in Baiga tribal populations, but in caste populations it varied from 11% (Dhimers) to 24% (Brahmins). Both tribal populations showed higher frequencies of allele *31 (17%-18%). For APOB, caste populations again showed bimodal distribution of alleles *35 and *37, but in tribal populations higher allele numbers (*47, *49) were also frequent. For YNZ22, extensive variation was observed for all populations studied. Allele *4 was the most common in caste populations, while alleles *2, *7, and *10 were prominent in tribal populations. The level of gene differentiation is not very high for the 3 systems studied in the 5 populations. Overall, allele frequency distribution, heterozygosity, and genetic diversity analysis show that the genetic diversity observed is socially and geographically structured.


Assuntos
DNA/análise , Etnicidade/genética , Variação Genética , Alelos , DNA/genética , Feminino , Frequência do Gene , Marcadores Genéticos , Genética Populacional , Humanos , Índia , Masculino , Biologia Molecular , Estudos de Amostragem , Classe Social , População Branca/genética
15.
Neurosci Lett ; 289(1): 29-32, 2000 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-10899401

RESUMO

Polyglutamine expansions in proteins are implicated in at least eight inherited neurodegenerative disorders, including Huntington's disease. These mutant proteins can form aggregates within the nucleus and processes of neurons possibly due to misfolding of the proteins. Polyglutamine aggregates are ubiquitinated and sequester molecular chaperone proteins and proteasome components. To investigate other protein components of polyglutamine aggregates, cerebral cortex and striata from patients with Huntington's disease and full-length cDNA transgenic mouse models for this disease were examined immunohistochemically for alpha-synuclein reactivity. Our findings demonstrate that alpha-synuclein can be used as a marker for huntingtin polyglutamine aggregates in both human and mice. Moreover in the HD transgenic mice, the intensity of immunoreactivity increases with age. The significance of recruitment of alpha-synuclein into huntingtin aggregates and its translocation away from the synapses remains to be determined. We propose that aberrant interaction of mutant huntingtin with other proteins, including alpha-synuclein, may influence disease progression.


Assuntos
Córtex Cerebral/química , Corpo Estriado/química , Doença de Huntington/metabolismo , Proteínas do Tecido Nervoso/análise , Proteínas Nucleares/análise , Peptídeos/análise , Fosfoproteínas/análise , Motivos de Aminoácidos , Animais , Córtex Cerebral/patologia , Corpo Estriado/patologia , Modelos Animais de Doenças , Feminino , Humanos , Proteína Huntingtina , Doença de Huntington/patologia , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/metabolismo , Fosfoproteínas/metabolismo , Dobramento de Proteína , Coelhos , Sinucleínas , alfa-Sinucleína
16.
Neurosci Lett ; 283(3): 233-5, 2000 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-10754231

RESUMO

Several neuroactive metabolites of the kynurenine pathway of tryptophan degradation have been speculatively linked to the pathophysiology of Huntington's Disease (HD). Here we demonstrate that the levels of two of these metabolites, the free radical generator 3-hydroxykynurenine (3HK) and the neuroprotectant kynurenate (KYNA), are increased in the neostriatum of stage 1 HD patients and in the brain of mice transgenic for full-length mutant huntingtin. In both cases, the elevation in 3HK was far more pronounced, resulting in significant increases in the 3HK/KYNA ratios. These data suggest that abnormal kynurenine pathway metabolism may play a role during the early phases of the neurodegenerative process in HD.


Assuntos
Doença de Huntington/genética , Doença de Huntington/metabolismo , Cinurenina/deficiência , Cinurenina/genética , Idoso , Animais , Modelos Animais de Doenças , Humanos , Proteína Huntingtina , Cinurenina/análogos & derivados , Cinurenina/biossíntese , Cinurenina/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Mutação/genética , Neocórtex/metabolismo , Neostriado/metabolismo , Proteínas do Tecido Nervoso/genética , Fármacos Neuroprotetores/metabolismo , Neurotoxinas/metabolismo , Proteínas Nucleares/genética
17.
Philos Trans R Soc Lond B Biol Sci ; 354(1386): 1035-45, 1999 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-10434303

RESUMO

Huntington's disease (HD) is a progressive neurodegenerative disorder characterized clinically by motor and psychiatric disturbances and pathologically by neuronal loss and gliosis (reactive astrocytosis) particularly in the striatum and cerebral cortex. We have recently created HD full-length cDNA transgenic mouse models that may serve as a paradigm for HD. A more detailed characterization of these models is presented here. The transgene encoding normal huntingtin consists of 9417 bp of the huntingtin coding sequences including 16 tandem CAGs coding for polyglutamines as part of exon 1. The transgene is driven by a heterologous cytomegalovirus promoter. Five independent transgenic mouse lines were obtained using this construct. An additional six transgenic lines were obtained using full-length HD constructs that have been modified to include either 48 or 89 CAG repeat expansions. Southern blot and densitometric analyses indicated unique integration sites for the transgene in each of the lines with a copy number ranging from two to 22 copies. Widespread expression of the transgene in brain, heart, spleen, kidney, lung, liver and gonads from each line was determined by Western blot analyses. In the brain, transgene expression was found in cerebral cortex, striatum, hippocampus and cerebellum. Expression of the transgene was as much as five times the endogenous mouse huntingtin level. Phenotypically, only mice expressing 48 or 89 CAG repeats manifested progressive behavioural and motor dysfunction. Early behavioural abnormalities were characterized by trunk curling and clasping of both fore- and hindlimbs when the animals were suspended by their tails. Subsequently, these mice exhibited hyperkinetic movements, including heightened exploratory activities, unidirectional rotational behaviour, backflipping and excessive grooming that lasted for several weeks. Eventually, the animals progressed to a hypokinetic phase consisting of slowed movements and lack of response to sensory stimuli. Urine retention or incontinence was also a prominent feature of the hypokinetic phase. At the end stage of the disease process, HD48(B,D) and HD89(A-C) mice became akinetic just prior to death. Neuropathological examination of mice at various stages indicated that it was only during the hypokinetic phase and thereafter when selective neuronal loss was most apparent. Regions of neurodegeneration and loss included the striatum, cerebral cortex, thalamus and hippocampus. TUNEL staining indicated an apoptotic mode of cell death in these brain regions. Comparative neuronal counts after Nissl staining showed as much as 20% loss of small and medium neurons in the striatum in mice at the hypokinetic and akinetic stages. Reactive astrocytosis accompanied the areas of neurodegeneration and loss. Polyglutamine aggregates in the form of neuronal intranuclear inclusions and diffuse nuclear and perinuclear aggregations were found in a small percentage of neurons, including those in brain regions that are typically spared in HD. This observation suggests that polyglutamine aggregates may not be sufficient to cause neuronal loss in HD. In both behavioural and neuropathological analyses, wild-type and transgenic animals with 16 CAG repeats were indistinguishable from each other and do not exhibit the changes observed for mice carrying the 48 and 89 CAG repeat mutations. Thus, animals expressing the CAG repeat expansions appear to represent clinically analogous models for HD pathogenesis, and may also provide insights into the underlying pathophysiological mechanisms of other triplet repeat disorders.


Assuntos
Encéfalo/patologia , Doença de Huntington/genética , Atividade Motora , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Proteínas Nucleares/genética , Animais , DNA Complementar , Éxons , Gliose , Humanos , Proteína Huntingtina , Doença de Huntington/patologia , Doença de Huntington/fisiopatologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/genética , Fenótipo , Expansão das Repetições de Trinucleotídeos/genética
18.
Trends Neurosci ; 22(6): 248-55, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10354600

RESUMO

Huntington's disease (HD) is an autosomal, dominantly inherited neurodegenerative disorder that is characterized by abnormal involuntary movements (chorea), intellectual impairment and selective neuronal loss. The expansion of a polymorphic trinucleotide repeat (the sequence CAG that codes for glutamine) to a length that exceeds 40 repeat units in exon 1 of the gene, HD, correlates with the onset and progression of the disease. The protein encoded by HD, huntingtin, is normally localized in the cytoplasm, whereas the mutant protein is also found in the nucleus, suggesting that its translocation to this site is important for the pathogenesis of HD. Although several proteins that interact with huntingtin have been identified in vitro, the significance of these interactions with the mutant protein in the pathogenesis of HD has yet to be determined. Recent progress in the development of cellular and animal models for the disease have provided invaluable insights and resources for studying the disease mechanisms underlying HD, and will be useful for screening and evaluating possible therapeutic strategies.


Assuntos
Encéfalo/fisiopatologia , Modelos Animais de Doenças , Doença de Huntington/genética , Biossíntese de Proteínas , Repetições de Trinucleotídeos/genética , Animais , Encéfalo/patologia , Dípteros , Progressão da Doença , Regulação da Expressão Gênica , Humanos , Doença de Huntington/patologia , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Mutação , Oligoquetos , Isoformas de Proteínas/genética , Translocação Genética
19.
J Neurosci ; 19(3): 964-73, 1999 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9920660

RESUMO

Neuronal intranuclear inclusions are found in the brains of patients with Huntington's disease and form from the polyglutamine-expanded N-terminal region of mutant huntingtin. To explore the properties of inclusions and their involvement in cell death, mouse clonal striatal cells were transiently transfected with truncated and full-length human wild-type and mutant huntingtin cDNAs. Both normal and mutant proteins localized in the cytoplasm, and infrequently, in dispersed and perinuclear vacuoles. Only mutant huntingtin formed nuclear and cytoplasmic inclusions, which increased with polyglutamine expansion and with time after transfection. Nuclear inclusions contained primarily cleaved N-terminal products, whereas cytoplasmic inclusions contained cleaved and larger intact proteins. Cells with wild-type or mutant protein had distinct apoptotic features (membrane blebbing, shrinkage, cellular fragmentation), but those with mutant huntingtin generated the most cell fragments (apoptotic bodies). The caspase inhibitor Z-VAD-FMK significantly increased cell survival but did not diminish nuclear and cytoplasmic inclusions. In contrast, Z-DEVD-FMK significantly reduced nuclear and cytoplasmic inclusions but did not increase survival. A series of N-terminal products was formed from truncated normal and mutant proteins and from full-length mutant huntingtin but not from full-length wild-type huntingtin. One prominent N-terminal product was blocked by Z-VAD-FMK. In summary, the formation of inclusions in clonal striatal cells corresponds to that seen in the HD brain and is separable from events that regulate cell death. N-terminal cleavage may be linked to mutant huntingtin's role in cell death.


Assuntos
Corpo Estriado/metabolismo , Mutação/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose/fisiologia , Western Blotting , Inibidores de Caspase , Sobrevivência Celular/fisiologia , Células Clonais , Corpo Estriado/citologia , Inibidores de Cisteína Proteinase/farmacologia , Humanos , Proteína Huntingtina , Corpos de Inclusão/fisiologia , Camundongos/embriologia , Neurônios/fisiologia , Oligopeptídeos/farmacologia , Distribuição Tecidual , Transfecção
20.
Nat Genet ; 20(2): 198-202, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9771716

RESUMO

Huntington disease (HD) is an adult-onset, autosomal dominant inherited human neurodegenerative disorder characterized by hyperkinetic involuntary movements, including motor restlessness and chorea, slowing of voluntary movements and cognitive impairment. Selective regional neuron loss and gliosis in striatum, cerebral cortex, thalamus, subthalamus and hippocampus are well recognized as neuropathological correlates for the clinical manifestations of HD. The underlying genetic mutation is the expansion of CAG trinucleotide repeats (coding for polyglutamines) to 36-121 copies in exon 1 of the HD gene. The HD mRNA and protein product (huntingtin) show widespread distribution, and thus much remains to be understood about the selective and progressive neurodegeneration in HD. To create an experimental animal model for HD, transgenic mice were generated showing widespread expression of full-length human HD cDNA with either 16, 48 or 89 CAG repeats. Only mice with 48 or 89 CAG repeats manifested progressive behavioural and motor dysfunction with neuron loss and gliosis in striatum, cerebral cortex, thalamus and hippocampus. These animals represent clinically relevant models for HD pathogenesis, and may provide insights into the underlying pathophysiological mechanisms of other triplet repeat disorders.


Assuntos
Comportamento Animal , Doença de Huntington/genética , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Proteínas Nucleares/genética , Animais , Apoptose , Corpo Estriado/química , DNA Complementar/biossíntese , DNA Complementar/química , Humanos , Proteína Huntingtina , Doença de Huntington/patologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/biossíntese , Proteínas Nucleares/análise , Proteínas Nucleares/biossíntese , Mapeamento por Restrição , Repetições de Trinucleotídeos/genética , Ubiquitinas/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...