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.
Biomolecules ; 9(8)2019 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-31344817

RESUMO

The role of mitochondria in Parkinson's disease (PD) has been investigated since the 1980s and is gaining attention with recent advances in PD genetics research. Mutations in PRKN and PTEN-Induced Putative Kinase 1 (PINK1) are well-established causes of autosomal recessive early-onset PD. Genetic and biochemical studies have revealed that PINK1 and Parkin proteins function together in the same biological pathway to govern mitochondrial quality control. These proteins have also been implicated in the regulation of innate and adaptive immunity and other mitochondrial functions. Additionally, structural studies on Parkin have delineated an activation mechanism and have identified druggable regions that are currently being explored by academic and industry groups. To de-risk therapeutic development for these genetic targets, The Michael J. Fox Foundation for Parkinson's Research (MJFF) has deployed a strategic funding and enabling framework that brings together the research community to discuss important breakthroughs and challenges in research on PINK1-Parkin biology, supports collaborative initiatives to further our understanding within this field and develops high-quality research tools and assays that are widely available to all researchers. The Foundation's efforts are leading to significant advances in understanding of the underlying biology of these genes, proteins and pathways and in the development of Parkinson's therapies.


Assuntos
Pesquisa Biomédica/economia , Fundações/organização & administração , Doença de Parkinson/genética , Proteínas Quinases/genética , Ubiquitina-Proteína Ligases/genética , Imunidade Adaptativa , Animais , Pesquisa Biomédica/organização & administração , Descoberta de Drogas , Apoio Financeiro , Humanos , Imunidade Inata , Mitocôndrias/metabolismo , Mitofagia , Terapia de Alvo Molecular , Mutação , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
2.
Nat Biotechnol ; 34(8): 838-44, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27376585

RESUMO

Rapid technological advances for the frequent monitoring of health parameters have raised the intriguing possibility that an individual's genotype could be predicted from phenotypic data alone. Here we used a machine learning approach to analyze the phenotypic effects of polymorphic mutations in a mouse model of Huntington's disease that determine disease presentation and age of onset. The resulting model correlated variation across 3,086 behavioral traits with seven different CAG-repeat lengths in the huntingtin gene (Htt). We selected behavioral signatures for age and CAG-repeat length that most robustly distinguished between mouse lines and validated the model by correctly predicting the repeat length of a blinded mouse line. Sufficient discriminatory power to accurately predict genotype required combined analysis of >200 phenotypic features. Our results suggest that autosomal dominant disease-causing mutations could be predicted through the use of subtle behavioral signatures that emerge in large-scale, combinatorial analyses. Our work provides an open data platform that we now share with the research community to aid efforts focused on understanding the pathways that link behavioral consequences to genetic variation in Huntington's disease.


Assuntos
Comportamento Animal , Genoma/genética , Proteína Huntingtina/genética , Doença de Huntington/genética , Camundongos/genética , Fenótipo , Animais , Mapeamento Cromossômico/métodos , Estudo de Associação Genômica Ampla/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Camundongos/classificação , Polimorfismo de Nucleotídeo Único/genética
3.
J Huntingtons Dis ; 3(2): 145-58, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25062858

RESUMO

BACKGROUND: Huntington's disease (HD) is characterized not only by severe motor deficits but also by early cognitive dysfunction that significantly increases the burden of the disease for patients and caregivers. Considerable efforts have concentrated, therefore, on the assessment of cognitive deficits in some HD mouse models. However, many of these models that exhibit cognitive deficits also have contemporaneous serious motor deficits, confounding interpretation of cognitive decline. OBJECTIVE: The BACHD and zQ175 mouse models present a more slowly progressing disease phenotype in both motor and cognitive domains, and might therefore offer a better opportunity to measure cognitive decline over a longer timeframe; such models could be useful in screening therapeutic compounds. In order to better define the cognitive impairments evident in BACHD and zQ175 HD mice, both were tested in an instrumental touchscreen visual discrimination assay designed to assess discrimination learning and cognitive flexibility. METHODS: BACHD and zQ175 mice, as well as their WT controls were tested for their ability to discriminate two complex visual stimuli. Following this discrimination phase, the reinforcement contingencies were reversed and the previously incorrect stimulus became the correct stimulus. In a final, third phase of testing, two novel stimuli were introduced and mice were required to undergo a second round of discrimination testing with these stimuli. RESULTS: Our results show that learning during the discrimination phase was similar between the WT and BACHD mice. In contrast, the zQ175 at 26 weeks of age showed decreased accuracy over the last 10 days of discrimination, compared to WT controls. During subsequent reversal and novel stimuli phases, both BACHD and zQ175 mice exhibited significant deficits compared to WT controls. CONCLUSIONS: Our results suggest that the BACHD, and for the first time, zQ175 HD models exhibit cognitive inflexibility and psychomotor slowing, a phenotype that is consistent with cognitive symptoms described in HD patients.


Assuntos
Transtornos Cognitivos/genética , Modelos Animais de Doenças , Doença de Huntington/genética , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Animais , Comportamento Animal/fisiologia , Transtornos Cognitivos/fisiopatologia , Computadores , Feminino , Técnicas de Introdução de Genes , Proteína Huntingtina , Doença de Huntington/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Desempenho Psicomotor/fisiologia , Tempo de Reação/genética , Reversão de Aprendizagem/fisiologia , Percepção Visual/fisiologia
4.
PLoS One ; 9(6): e99520, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24955833

RESUMO

Huntington's disease (HD) is an autosomal dominant, progressive neurodegenerative disorder caused by expansion of CAG repeats in the huntingtin gene. Tissue transglutaminase 2 (TG2), a multi-functional enzyme, was found to be increased both in HD patients and in mouse models of the disease. Furthermore, beneficial effects have been reported from the genetic ablation of TG2 in R6/2 and R6/1 mouse lines. To further evaluate the validity of this target for the treatment of HD, we examined the effects of TG2 deletion in two genetic mouse models of HD: R6/2 CAG 240 and zQ175 knock in (KI). Contrary to previous reports, under rigorous experimental conditions we found that TG2 ablation had no effect on either motor or cognitive deficits, or on the weight loss. In addition, under optimal husbandry conditions, TG2 ablation did not extend R6/2 lifespan. Moreover, TG2 deletion did not change the huntingtin aggregate load in cortex or striatum and did not decrease the brain atrophy observed in either mouse line. Finally, no amelioration of the dysregulation of striatal and cortical gene markers was detected. We conclude that TG2 is not a valid therapeutic target for the treatment of HD.


Assuntos
Proteínas de Ligação ao GTP/genética , Deleção de Genes , Doença de Huntington/enzimologia , Doença de Huntington/patologia , Transglutaminases/genética , Animais , Atrofia , Comportamento Animal , Encéfalo/metabolismo , Encéfalo/patologia , Transtornos Cognitivos/complicações , Cruzamentos Genéticos , Discriminação Psicológica , Modelos Animais de Doenças , Feminino , Genótipo , Doença de Huntington/complicações , Ligantes , Masculino , Aprendizagem em Labirinto , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes Neurológicos , Fenótipo , Proteína 2 Glutamina gama-Glutamiltransferase , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sobrevida , Redução de Peso
5.
PLoS Curr ; 52013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-24042107

RESUMO

The genome of the Bacterial Artificial Chromosome (BAC) transgenic mouse model of Huntington's Disease (BAC HD) contains the 170 kb human HTT locus modified by the addition of exon 1 with 97 mixed CAA-CAG repeats. BAC HD mice present robust behavioral deficits in both the open field and the accelerating rotarod tests, two standard behavioral assays of motor function. BAC HD mice, however, also typically present significantly increased body weights relative to wildtype littermate controls (WT) which potentially confounds the interpretation of any motor deficits associated directly with the effects of mutant huntingtin. In order to evaluate this possible confound of body weight, we directly compared the performance of BAC HD and WT female mice under food restricted versus free feeding conditions in both the open field and rotarod tasks to test the hypothesis that some of the motor deficits observed in this HTT-transgenic mouse line results solely from increased body weight. Our results suggest that the rotarod deficit exhibited by BAC HD mice is modulated by both body weight and non-body weight factors resulting from overexpression of full length mutant Htt. When body weights of WT and BAC HD transgenic mice were normalized using restricted feeding, the deficits exhibited by BAC HD mice on the rotarod task were less marked, but were still significant. Since the rotarod deficit between WT and BAC HD mice is attenuated when body weight is normalized by food restriction, utilization of this task in BAC HD mice during pre-clinical evaluation must be powered accordingly and results carefully considered as therapeutic benefit can result from decreased overall body weight and or motoric improvement that may not be related to body mass. Furthermore, after controlling for body weight differences, the hypoactive phenotype displayed by ad libitum fed BAC HD mice in the open field assay was not observed in the BAC HD mice undergoing food restriction. These findings suggest that assessment of spontaneous locomotor activity, as measured in the open field test, may not be the appropriate behavioral endpoint to evaluate the BAC HD mouse during preclinical evaluation since it appears that the apparent hypoactive phenotype in this model is driven primarily by body weight differences.

6.
J Neurosci ; 32(19): 6456-67, 2012 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-22573668

RESUMO

A hallmark feature of Huntington's disease pathology is the atrophy of brain regions including, but not limited to, the striatum. Though MRI studies have identified structural CNS changes in several Huntington's disease (HD) mouse models, the functional consequences of HD pathology during the progression of the disease have yet to be investigated using in vivo functional MRI (fMRI). To address this issue, we first established the structural and functional MRI phenotype of juvenile HD mouse model R6/2 at early and advanced stages of disease. Significantly higher fMRI signals [relative cerebral blood volumes (rCBVs)] and atrophy were observed in both age groups in specific brain regions. Next, fMRI results were correlated with electrophysiological analysis, which showed abnormal increases in neuronal activity in affected brain regions, thus identifying a mechanism accounting for the abnormal fMRI findings. [(14)C] 2-deoxyglucose maps to investigate patterns of glucose utilization were also generated. An interesting mismatch between increases in rCBV and decreases in glucose uptake was observed. Finally, we evaluated the sensitivity of this mouse line to audiogenic seizures early in the disease course. We found that R6/2 mice had an increased susceptibility to develop seizures. Together, these findings identified seizure activity in R6/2 mice and show that neuroimaging measures sensitive to oxygen metabolism can be used as in vivo biomarkers, preceding the onset of an overt behavioral phenotype. Since fMRI-rCBV can also be obtained in patients, we propose that it may serve as a translational tool to evaluate therapeutic responses in humans and HD mouse models.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Progressão da Doença , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Convulsões/metabolismo , Convulsões/patologia , Animais , Feminino , Predisposição Genética para Doença , Doença de Huntington/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Consumo de Oxigênio/fisiologia , Convulsões/etiologia , Fatores de Tempo
7.
PLoS One ; 7(12): e49838, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23284626

RESUMO

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterized by motor, cognitive and psychiatric manifestations. Since the mutation responsible for the disease was identified as an unstable expansion of CAG repeats in the gene encoding the huntingtin protein in 1993, numerous mouse models of HD have been generated to study disease pathogenesis and evaluate potential therapeutic approaches. Of these, knock-in models best mimic the human condition from a genetic perspective since they express the mutation in the appropriate genetic and protein context. Behaviorally, however, while some abnormal phenotypes have been detected in knock-in mouse models, a model with an earlier and more robust phenotype than the existing models is required. We describe here for the first time a new mouse line, the zQ175 knock-in mouse, derived from a spontaneous expansion of the CAG copy number in our CAG 140 knock-in colony [1]. Given the inverse relationship typically observed between age of HD onset and length of CAG repeat, since this new mouse line carries a significantly higher CAG repeat length it was expected to be more significantly impaired than the parent line. Using a battery of behavioral tests we evaluated both heterozygous and homozygous zQ175 mice. Homozygous mice showed motor and grip strength abnormalities with an early onset (8 and 4 weeks of age, respectively), which were followed by deficits in rotarod and climbing activity at 30 weeks of age and by cognitive deficits at around 1 year of age. Of particular interest for translational work, we also found clear behavioral deficits in heterozygous mice from around 4.5 months of age, especially in the dark phase of the diurnal cycle. Decreased body weight was observed in both heterozygotes and homozygotes, along with significantly reduced survival in the homozygotes. In addition, we detected an early and significant decrease of striatal gene markers from 12 weeks of age. These data suggest that the zQ175 knock-in line could be a suitable model for the evaluation of therapeutic approaches and early events in the pathogenesis of HD.


Assuntos
Comportamento Animal , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Doença de Huntington/genética , Animais , Comportamento Animal/efeitos da radiação , Peso Corporal/genética , Cognição/fisiologia , Escuridão , Feminino , Marcadores Genéticos/genética , Força da Mão/fisiologia , Heterozigoto , Homozigoto , Doença de Huntington/fisiopatologia , Masculino , Camundongos , Neostriado/metabolismo , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sequências Repetitivas de Ácido Nucleico , Teste de Desempenho do Rota-Rod , Análise de Sobrevida , Transcrição Gênica/genética
8.
PLoS One ; 5(3): e9793, 2010 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-20339553

RESUMO

Previous studies of the effects of coenzyme Q10 and minocycline on mouse models of Huntington's disease have produced conflicting results regarding their efficacy in behavioral tests. Using our recently published best practices for husbandry and testing for mouse models of Huntington's disease, we report that neither coenzyme Q10 nor minocycline had significant beneficial effects on measures of motor function, general health (open field, rotarod, grip strength, rearing-climbing, body weight and survival) in the R6/2 mouse model. The higher doses of minocycline, on the contrary, reduced survival. We were thus unable to confirm the previously reported benefits for these two drugs, and we discuss potential reasons for these discrepancies, such as the effects of husbandry and nutrition.


Assuntos
Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Doença de Huntington/genética , Minociclina/farmacologia , Ubiquinona/análogos & derivados , Animais , Antibacterianos/farmacologia , Peso Corporal/efeitos dos fármacos , Feminino , Força da Mão , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Destreza Motora/efeitos dos fármacos , Ubiquinona/farmacologia
9.
NeuroRx ; 2(3): 465-70, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16389309

RESUMO

Huntington's disease is an autosomal dominant neurodegenerative disorder that is characterized by motor, cognitive, and psychiatric alterations. The mutation responsible for this fatal disease is an abnormally expanded and unstable CAG repeat within the coding region of the gene encoding huntingtin. Numerous mouse models have been generated that constitute invaluable tools to examine the pathogenesis of the disease and to develop and evaluate novel therapies. Among those models, knock-in mice provide a genetically precise reproduction of the human condition. The slow progression and early development of behavioral, pathological, cellular, and molecular abnormalities in knock-in mice make these animals valuable to understand the early pathological events triggered by the mutation. This review describes the different knock-in models generated, the insight gained from them, and their value in the development and testing of prospective treatments of the disease.


Assuntos
Doença de Huntington/genética , Doença de Huntington/patologia , Camundongos Transgênicos/genética , Animais , Comportamento Animal/fisiologia , Encéfalo/patologia , Humanos , Doença de Huntington/psicologia , Camundongos , Neurônios/patologia
10.
J Comp Neurol ; 465(1): 11-26, 2003 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-12926013

RESUMO

Huntington's disease (HD) is caused by an abnormal expansion of CAG repeats in the gene encoding huntingtin. The development of therapies for HD requires preclinical testing of drugs in animal models that reproduce the dysfunction and regionally specific pathology observed in HD. We have developed a new knock-in mouse model of HD with a chimeric mouse/human exon 1 containing 140 CAG repeats inserted in the murine huntingtin gene. These mice displayed an increased locomotor activity and rearing at 1 month of age, followed by hypoactivity at 4 months and gait anomalies at 1 year. Behavioral symptoms preceded neuropathological anomalies, which became intense and widespread only at 4 months of age. These consisted of nuclear staining for huntingtin and huntingtin-containing nuclear and neuropil aggregates that first appeared in the striatum, nucleus accumbens, and olfactory tubercle. Interestingly, regions with early pathology all receive dense dopaminergic inputs, supporting accumulating evidence for a role of dopamine in HD pathology. Nuclear staining and aggregates predominated in striatum and layer II/III and deep layer V of the cerebral cortex, whereas neuropil aggregates were found in the globus pallidus and layer IV/superficial layer V of the cerebral cortex. The olfactory system displayed early and marked aggregate accumulation, which may be relevant to the early deficit in odor discrimination observed in patients with HD. Because of their early behavioral anomalies and regionally specific pathology, these mice provide a powerful tool with which to evaluate the effectiveness of new therapies and to study the mechanisms involved in the neuropathology of HD.


Assuntos
Encéfalo/patologia , Encéfalo/fisiopatologia , Doença de Huntington/patologia , Doença de Huntington/fisiopatologia , Proteínas do Tecido Nervoso/deficiência , Neurônios/patologia , Proteínas Nucleares/deficiência , Repetições de Trinucleotídeos/genética , Animais , Encéfalo/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Feminino , Asseio Animal/fisiologia , Proteína Huntingtina , Doença de Huntington/genética , Imuno-Histoquímica , Corpos de Inclusão/genética , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Coxeadura Animal/genética , Coxeadura Animal/patologia , Coxeadura Animal/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/genética , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Neurópilo/metabolismo , Neurópilo/patologia , Proteínas Nucleares/genética , Núcleo Accumbens/metabolismo , Núcleo Accumbens/patologia , Núcleo Accumbens/fisiopatologia , Condutos Olfatórios/metabolismo , Condutos Olfatórios/patologia , Condutos Olfatórios/fisiopatologia , Proteínas Recombinantes de Fusão/genética , Fatores de Tempo
11.
J Neurosci ; 22(18): 8266-76, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12223581

RESUMO

Huntington's disease (HD) is characterized by a progressive loss of neurons in the striatum and cerebral cortex and is caused by a CAG repeat expansion in the gene encoding huntingtin. Mice with the mutation inserted into their own huntingtin gene (knock-in mice) are, genetically, the best models of the human disease. Here we show for the first time that knock-in mice with 94 CAG repeats develop a robust and early motor phenotype at 2 months of age, characterized by increased rearing at night. This initial increase in repetitive movements was followed by decreased locomotion at 4 and 6 months, despite a normal life span. The decrease in striatal enkephalin mRNA that is known to occur at 4 months was not present at 2 months, when increased rearing was observed. Both the hyperactive and hypoactive phases of motor dysfunction preceded the detection of nuclear microaggregates of mutated huntingtin in striatal neurons. Nuclear microaggregates, defined as small huntingtin-positive punctas detected by light microscopy, were very rare at 4 months but became widely distributed in striatal neurons at 6 months. Nuclear inclusions did not appear until 18 months. When present, nuclear microaggregates predominated in the striosomal compartment of the striatum, providing a possible explanation for the different neuronal vulnerability of striatal compartments observed in humans. The early motor phenotype observed in the knock-in mouse is reminiscent of repetitive movements often observed in early HD and provides a novel opportunity to assess the ability of therapies to prevent the initial effects of the mutation in vivo.


Assuntos
Corpo Estriado/metabolismo , Doença de Huntington/fisiopatologia , Corpos de Inclusão/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Animais , Comportamento Animal , Contagem de Células , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Corpo Estriado/química , Corpo Estriado/patologia , Modelos Animais de Doenças , Progressão da Doença , Encefalinas/biossíntese , Encefalinas/genética , Feminino , Asseio Animal , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/patologia , Corpos de Inclusão/ultraestrutura , Substâncias Macromoleculares , Masculino , Camundongos , Camundongos Mutantes , Atividade Motora , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Neurônios/patologia , Proteínas Nucleares/genética , Especificidade de Órgãos , Fenótipo , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Receptores Opioides mu/metabolismo , Expansão das Repetições de Trinucleotídeos/genética
12.
Trends Pharmacol Sci ; 23(1): 32-9, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11804649

RESUMO

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder. In 1993 the mutation that causes HD was identified as an unstable expansion of CAG repeats in the IT15 gene. Since then one of the most important advances in HD research has been the generation of various mouse models that enable the exploration of early pathological, molecular and cellular abnormalities produced by the mutation. In addition, these models have made it possible to test different pharmacological approaches to delay the onset or slow the progression of HD. In this article, insights gained from mouse models towards the understanding of HD and the design of new therapeutic strategies are discussed.


Assuntos
Doença de Huntington/genética , Modelos Animais , Animais , Creatina/uso terapêutico , Ácido Dicloroacético/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Humanos , Proteína Huntingtina , Doença de Huntington/tratamento farmacológico , Doença de Huntington/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Minociclina/uso terapêutico , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Proteínas/genética , Expansão das Repetições de Trinucleotídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...