Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
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.
Neuroscience ; 409: 169-179, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31029729

RESUMO

Parkinson's disease (PD) is the most common neurodegenerative movement disorder and is characterized by the loss of neurons in the substantia nigra that project to the striatum and release dopamine (DA), which is required for normal movement. Common non-motor symptoms likely involve abnormalities with other neurotransmitters, such as serotonin, norepinephrine, acetylcholine, glycine, glutamate and gamma-aminobutyric acid (GABA). As part of a broad effort to provide better PD research tools, the Michael J. Fox Foundation for Parkinson's Research funded the generation and characterization of knockout (KO) rats for genes with PD-linked mutations, including PINK1, Parkin, DJ-1 and LRRK2. Here we extend the phenotypic characterization of these lines of KO rats to include in vivo microdialysis to measure both basal and potassium-induced release of the above neurotransmitters and their metabolites in the striatum of awake and freely moving rats at ages 4, 8 and 12 months compared to wild-type (WT) rats. We found age-dependent abnormalities in basal DA, glutamate and acetylcholine in PINK1 KO rats and age-dependent abnormalities in basal DA metabolites in Parkin and LRRK2 KO rats. Parkin KO rats had increased glycine release while DJ-1 KO rats had decreased glutamate release and increased acetylcholine release compared to WT rats. All lines except DJ-1 KO rats showed age-dependent changes in release of one or more neurotransmitters. Our data suggest these rats may be useful for studies of PD-related synaptic dysfunction and neurotransmitter dynamics as well as studies of the normal and pathogenic functions of these genes with PD-linked mutations.


Assuntos
Acetilcolina/metabolismo , Encéfalo/metabolismo , Dopamina/metabolismo , Ácido Glutâmico/metabolismo , Doença de Parkinson/metabolismo , Animais , Neurônios Dopaminérgicos/metabolismo , Técnicas de Inativação de Genes , Glicina/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Masculino , Doença de Parkinson/genética , Proteína Desglicase DJ-1/genética , Proteína Desglicase DJ-1/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Ratos , Serotonina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ácido gama-Aminobutírico/metabolismo
3.
J Parkinsons Dis ; 9(1): 31-61, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30400107

RESUMO

The convergence of human molecular genetics and Lewy pathology of Parkinson's disease (PD) have led to a robust, clinical-stage pipeline of alpha-synuclein (α-syn)-targeted therapies that have the potential to slow or stop the progression of PD and other synucleinopathies. To facilitate the development of these and earlier stage investigational molecules, the Michael J. Fox Foundation for Parkinson's Research convened a group of leaders in the field of PD research from academia and industry, the Alpha-Synuclein Clinical Path Working Group. This group set out to develop recommendations on preclinical and clinical research that can de-risk the development of α-syn targeting therapies. This consensus white paper provides a translational framework, from the selection of animal models and associated end-points to decision-driving biomarkers as well as considerations for the design of clinical proof-of-concept studies. It also identifies current gaps in our biomarker toolkit and the status of the discovery and validation of α-syn-associated biomarkers that could help fill these gaps. Further, it highlights the importance of the emerging digital technology to supplement the capture and monitoring of clinical outcomes. Although the development of disease-modifying therapies targeting α-syn face profound challenges, we remain optimistic that meaningful strides will be made soon toward the identification and approval of disease-modifying therapeutics targeting α-syn.


Assuntos
Biomarcadores , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Guias como Assunto , Doença de Parkinson/tratamento farmacológico , Estudo de Prova de Conceito , Pesquisa Translacional Biomédica , alfa-Sinucleína/efeitos dos fármacos , Animais , Consenso , Humanos , Doença de Parkinson/diagnóstico , Projetos de Pesquisa
4.
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
5.
Mov Disord ; 29(11): 1375-90, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25216369

RESUMO

Mouse models of Huntington's disease (HD) recapitulate many aspects of the human disease. These genetically modified mice are powerful tools that are used not only to examine the pathogenesis of the disease, but also to assess the efficacy of potential new treatments. Disappointingly, in the past few years we have seen the success of potential therapies in animal studies, subsequently followed by failure in clinical trials. We discuss here a number of factors that influence the translatability of findings from the preclinical to the clinical realm. In particular, we discuss issues related to sample size, reporting of information regarding experimental protocols and mouse models, assignment to experimental groups, incorporation of cognitive measures for early phases of the disease, environmental enrichment, surrogate measures for survival, and the use of more than one HD mouse model. Although it is reasonable to question the appropriateness of the animal models used, we argue that it is more parsimonious to assume that improvements in experimental design and publication of negative results will lead to improved translatability to the clinic and insights about HD pathophysiology.


Assuntos
Modelos Animais de Doenças , Doença de Huntington/terapia , Pesquisa Translacional Biomédica/métodos , Animais , Transtornos Cognitivos/etiologia , Humanos , Doença de Huntington/complicações , Camundongos
6.
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
7.
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
8.
Hum Mol Genet ; 23(11): 2995-3007, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24436303

RESUMO

Protein acetylation, which is central to transcriptional control as well as other cellular processes, is disrupted in Huntington's disease (HD). Treatments that restore global acetylation levels, such as inhibiting histone deacetylases (HDACs), are effective in suppressing HD pathology in model organisms. However, agents that selectively target the disease-relevant HDACs have not been available. SirT1 (Sir2 in Drosophila melanogaster) deacetylates histones and other proteins including transcription factors. Genetically reducing, but not eliminating, Sir2 has been shown to suppress HD pathology in model organisms. To date, small molecule inhibitors of sirtuins have exhibited low potency and unattractive pharmacological and biopharmaceutical properties. Here, we show that highly selective pharmacological inhibition of Drosophila Sir2 and mammalian SirT1 using the novel inhibitor selisistat (selisistat; 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide) can suppress HD pathology caused by mutant huntingtin exon 1 fragments in Drosophila, mammalian cells and mice. We have validated Sir2 as the in vivo target of selisistat by showing that genetic elimination of Sir2 eradicates the effect of this inhibitor in Drosophila. The specificity of selisistat is shown by its effect on recombinant sirtuins in mammalian cells. Reduction of HD pathology by selisistat in Drosophila, mammalian cells and mouse models of HD suggests that this inhibitor has potential as an effective therapeutic treatment for human disease and may also serve as a tool to better understand the downstream pathways of SirT1/Sir2 that may be critical for HD.


Assuntos
Carbazóis/administração & dosagem , Proteínas de Drosophila/antagonistas & inibidores , Inibidores Enzimáticos/administração & dosagem , Doença de Huntington/tratamento farmacológico , Doença de Huntington/enzimologia , Sirtuína 1/antagonistas & inibidores , Sirtuínas/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Doença de Huntington/genética , Doença de Huntington/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células PC12 , Ratos , Ratos Sprague-Dawley , Sirtuína 1/genética , Sirtuína 1/metabolismo , Sirtuínas/genética , Sirtuínas/metabolismo
9.
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.

10.
PLoS Curr ; 52013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23863947

RESUMO

Phenotyping with traditional behavioral assays constitutes a major bottleneck in the primary screening, characterization, and validation of genetic mouse models of disease, leading to downstream delays in drug discovery efforts. We present a novel and comprehensive one-stop approach to phenotyping, the PhenoCube™. This system simultaneously captures the cognitive performance, motor activity, and circadian patterns of group-housed mice by use of home-cage operant conditioning modules (IntelliCage) and custom-built computer vision software. We evaluated two different mouse models of Huntington's Disease (HD), the R6/2 and the BACHD in the PhenoCube™ system. Our results demonstrated that this system can efficiently capture and track alterations in both cognitive performance and locomotor activity patterns associated with these disease models. This work extends our prior demonstration that PhenoCube™ can characterize circadian dysfunction in BACHD mice and shows that this system, with the experimental protocols used, is a sensitive and efficient tool for a first pass high-throughput screening of mouse disease models in general and mouse models of neurodegeneration in particular.

11.
Neurobiol Dis ; 52: 84-93, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23220414

RESUMO

Although the huntingtin gene is expressed in brain throughout life, phenotypically Huntington's disease (HD) begins only in midlife and affects specific brain regions. Here, to investigate regional vulnerability in the disease, we used functional magnetic resonance imaging (fMRI) to translationally link studies in patients with a mouse model of disease. Using fMRI, we mapped cerebral blood volume (CBV) in three groups: HD patients, symptom-free carriers of the huntingtin genetic mutation, and age-matched controls. In contrast to a region in the anterior caudate, in which dysfunction was linked to genotype independent of phenotype, a region in the posterior body of the caudate was differentially associated with disease phenotype. Guided by these observations, we harvested regions from the anterior and posterior body of the caudate in postmortem control and HD human brain tissue. Gene-expression profiling identified two molecules whose expression levels were most strongly correlated with regional vulnerability - protein phosphatase 1 regulatory subunit 7 (PPP1R7) and Wnt inhibitory factor-1 (WIF-1). To verify and potentially extend these findings, we turned to the YAC128 (C57BL/6J) HD transgenic mice. By fMRI we longitudinally mapped CBV in transgenic and wildtype (WT) mice, and over time, abnormally low fMRI signal emerged selectively in the dorsal striatum. A relatively unaffected brain region, primary somatosensory cortex (S1), was used as a control. Both dorsal striatum and S1 were harvested from transgenic and WT mice and molecular analysis confirmed that PPP1R7 deficiency was strongly correlated with the phenotype. Together, converging findings in human HD patients and this HD mouse model suggest a functional pattern of caudate vulnerability and that variation in expression levels of herein identified molecules correlate with this pattern of vulnerability.


Assuntos
Circulação Cerebrovascular/fisiologia , Corpo Estriado/fisiopatologia , Doença de Huntington/fisiopatologia , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Idoso , Animais , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/metabolismo , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Mutação , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Proteínas Nucleares/metabolismo , Fenótipo , Proteína Fosfatase 1/genética , Proteína Fosfatase 1/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
12.
J Neurosci ; 32(22): 7454-65, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22649225

RESUMO

Huntington's disease (HD) is caused by a mutation in the huntingtin (htt) gene encoding an expansion of glutamine repeats at the N terminus of the Htt protein. Proteolysis of Htt has been identified as a critical pathological event in HD models. In particular, it has been postulated that proteolysis of Htt at the putative caspase-6 cleavage site (at amino acid Asp-586) plays a critical role in disease progression and pathogenesis. However, whether caspase-6 is indeed the essential enzyme that cleaves Htt at this site in vivo has not been determined. To evaluate, we crossed the BACHD mouse model with a caspase-6 knock-out mouse (Casp6(-/-)). Western blot and immunocytochemistry confirmed the lack of caspase-6 protein in Casp6(-/-) mice, regardless of HD genotype. We predicted the Casp6(-/-) mouse would have reduced levels of caspase-6 Htt fragments and increased levels of full-length Htt protein. In contrast, we found a significant reduction of full-length mutant Htt (mHtt) and fragments in the striatum of BACHD Casp6(-/-) mice. Importantly, we detected the presence of Htt fragments consistent with cleavage at amino acid Asp-586 of Htt in the BACHD Casp6(-/-) mouse, indicating that caspase-6 activity cannot fully account for the generation of the Htt 586 fragment in vivo. Our data are not consistent with the hypothesis that caspase-6 activity is critical in generating a potentially toxic 586 aa Htt fragment in vivo. However, our studies do suggest a role for caspase-6 activity in clearance pathways for mHtt protein.


Assuntos
Ácido Aspártico/metabolismo , Caspase 6/metabolismo , Regulação da Expressão Gênica/genética , Doença de Huntington/metabolismo , Doença de Huntington/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Fatores Etários , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Ácido Aspártico/genética , Peso Corporal/genética , Encéfalo/metabolismo , Encéfalo/patologia , Caspase 6/deficiência , Células Cultivadas , Corpo Estriado/citologia , Modelos Animais de Doenças , Embrião de Mamíferos , Comportamento Exploratório/fisiologia , Feminino , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/patologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Atividade Motora/genética , Proteínas do Tecido Nervoso/genética , Neurônios , Proteólise , RNA Interferente Pequeno/metabolismo , Teste de Desempenho do Rota-Rod , Expansão das Repetições de Trinucleotídeos/genética , Ubiquitinação/genética
13.
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
14.
PLoS Curr ; 4: RRN1303, 2012 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-22367489

RESUMO

Homozygosis for the rd1 mutation in the Pbe6b gene results in the loss of the rod beta-subunit of the cyclic GMP phosphodiesterase and, eventually, of all rod and cone photoreceptors. The R6/2 mouse line is a widely used model of Huntington's disease (HD). The original line was made available on a mixed background obtained by crossing, via ovarian transplant, female R6/2 (on a B6CBA mixed background) with male B6CBAF1/J mice. As the CBA/J strain used in the US is homozygous for the rd1 mutation and the breeding scheme does not ensure heterozygosis for the mutation, a significant percentage of the offspring on this mixed background is expected to be homozygous for the rd1 mutation. We investigate here the effect of rd1 homozygosis on motor function and examined the effects of the mutation on the R6/2 phenotype. Homozygosis for the rd1 mutation resulted in increased activity in the open field test and reduced rotarod test performance. In addition, rd1 mutation absence or heterozygosis reduced the differences between the R6/2 and the WT mice. Our recommendation for the neurodegeneration field, and for all mouse studies in general, is to carefully control homozygosis for retinal degeneration mutation, even when using tests of motor function.

15.
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
16.
PLoS Curr ; 3: RRN1225, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21479110

RESUMO

Huntington's disease (HD) is a progressive neurodegenerative disease marked by psychiatric and motor problems. Recently, these findings have been extended to deficits in sleep and circadian function that can be observed in HD patients and in HD mouse models, with abnormal sleep patterns correlating with symptom severity in patients. Here, we studied the behavior of the BAC HD mouse model using an 24/7 automated system; the results indicate significant lengthening of the circadian period in the mutant mice. These results reinforce previous findings in HD models and symptomatic HD patients, indicating that circadian dysfunction is a core feature of HD.

17.
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
18.
Neurobiol Dis ; 35(3): 319-36, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19464370

RESUMO

Huntington's disease (HD) is one of the few neurodegenerative diseases with a known genetic cause, knowledge that has enabled the creation of animal models using genetic manipulations that aim to recapitulate HD pathology. The study of behavioral and neuropathological phenotypes of these HD models, however, has been plagued by inconsistent results across laboratories stemming from the lack of standardized husbandry and testing conditions, in addition to the intrinsic differences between the models. We have compared different HD models using standardized conditions to identify the most robust phenotypic differences, best suited for preclinical therapeutic efficacy studies. With a battery of tests of sensory-motor function, such as the open field and prepulse inhibition tests, we replicate previous results showing a strong and progressive behavioral deficit in the R6/2 line with an average of 129 CAG repeats in a mixed CBA/J and C57BL/6J background. We present the first behavioral characterization of a new model, an R6/2 line with an average of 248 CAG repeats in a pure C57BL/6J background, which also showed a progressive and robust phenotype. The BACHD in a FVB/N background showed robust and progressive behavioral phenotype, while the YAC128 full-length model on either an FVB/N or a C57BL/6J background generally showed milder deficits. Finally, the Hdh(Q111) knock-in mouse on a CD1 background showed very mild deficits. This first extensive standardized cross-characterization of several HD animal models under standardized conditions highlights several behavioral outcomes, such as hypoactivity, amenable to standardized preclinical therapeutic drug screening.


Assuntos
Comportamento Animal , Modelos Animais de Doenças , Doença de Huntington , Atividade Motora , Envelhecimento , Animais , Feminino , Técnicas de Introdução de Genes , Genótipo , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/genética , Proteínas do Tecido Nervoso/genética , Testes Neuropsicológicos , Proteínas Nucleares/genética , Fenótipo , Caracteres Sexuais
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...