Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Neurobiol Dis ; 134: 104614, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31605779

RESUMO

The G2019S substitution in the kinase domain of LRRK2 (LRRK2G2019S) is the most prevalent mutation associated with Parkinson's disease (PD). Neurotoxic effects of LRRK2G2019S are thought to result from an increase in its kinase activity as compared to wild type LRRK2. However, it is unclear whether the kinase domain of LRRK2G2019S is sufficient to trigger degeneration or if the full length protein is required. To address this question, we generated constructs corresponding to the C-terminal domain of LRRK2 (ΔLRRK2). A kinase activity that was increased by G2019➔S substitution could be detected in ΔLRRK2. However biochemical experiments suggested it did not bind or phosphorylate the substrate RAB10, in contrast to full length LRRK2. The overexpression of ΔLRRK2G2019S in the rat striatum using lentiviral vectors (LVs) offered a straightforward and simple way to investigate its effects in neurons in vivo. Results from a RT-qPCR array analysis indicated that ΔLRRK2G2019S led to significant mRNA expression changes consistent with a kinase-dependent mechanism. We next asked whether ΔLRRK2 could be sufficient to trigger neurodegeneration in the substantia nigra pars compacta (SNc) in adult rats. Six months after infection of the substantia nigra pars compacta (SNc) with LV-ΔLRRK2WT or LV-ΔLRRK2G2019S, the number of DA neurons was unchanged. To examine whether higher levels of ΔLRRK2G2019S could trigger degeneration we cloned ΔLRRK2 in AAV2/9 construct. As expected, AAV2/9 injected in the SNc led to neuronal expression of ΔLRRK2WT and ΔLRRK2G2019S at much higher levels than those obtained with LVs. Six months after injection, unbiased stereology showed that AAV-ΔLRRK2G2019S produced a significant ~30% loss of neurons positive for tyrosine hydroxylase- and for the vesicular dopamine transporter whereas AAV-ΔLRRK2WT did not. These findings show that overexpression of the C-terminal part of LRRK2 containing the mutant kinase domain is sufficient to trigger degeneration of DA neurons, through cell-autonomous mechanisms, possibly independent of RAB10.


Assuntos
Neurônios Dopaminérgicos/patologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Degeneração Neural/genética , Doença de Parkinson , Domínios Proteicos/genética , Animais , Técnicas de Transferência de Genes , Vetores Genéticos , Células HEK293 , Humanos , Lentivirus , Masculino , Mutação , Degeneração Neural/patologia , Parte Compacta da Substância Negra , Ratos , Ratos Sprague-Dawley
2.
Brain ; 141(2): 535-549, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29253129

RESUMO

Tauopathies are neurodegenerative diseases characterized by the aggregation of tau protein. These pathologies exhibit a wide variety of clinical and anatomo-pathological presentations, which may result from different pathological mechanisms. Although tau inclusions are a common feature in all these diseases, recent evidence instead implicates small oligomeric aggregates as drivers of tau-induced toxicity. Hence in vivo model systems displaying either soluble or fibrillary forms of wild-type or mutant tau are needed to better identify their respective pathological pathways. Here we used adeno-associated viruses to mediate gene transfer of human tau to the rat brain to develop models of pure tauopathies. Two different constructs were used, each giving rise to a specific phenotype developing in less than 3 months. First, hTAUWT overexpression led to a strong hyperphosphorylation of the protein, which was associated with neurotoxicity in the absence of any significant aggregation. In sharp contrast, its co-expression with the pro-aggregation peptide TauRD-ΔK280 in the hTAUProAggr group strongly promoted its aggregation into Gallyas-positive neurofibrillary tangles, while preserving neuronal survival. Our results support the hypothesis that soluble tau species are key players of tau-induced neurodegeneration.


Assuntos
Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Coloração pela Prata , Tauopatias/diagnóstico por imagem , Transdução Genética , Vimentina/metabolismo , Proteínas tau/genética
3.
Prog Brain Res ; 230: 283-303, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28552233

RESUMO

Stem cell-based therapy trials for the treatment of neurodegenerative diseases such as Parkinson's and Huntington's disease are being actively prepared both at the preclinical and clinical level. Preclinical validation of these stem cell transplantations necessitates to implement a translational continuum to take one pluripotent stem cell through the point of "first-in-man" clinical trial. Main steps along this translational continuum include stem cell GMP production, in vitro optimization of differentiation protocols necessary to direct stem cells to the desired neuronal phenotype, and evaluation of functional efficacy in animal models including large animal models. Whereas the first two steps only require in vitro techniques and can be expedited smoothly, the last step is generally time consuming as functional assessment requires transplantation of the animal models with sufficient time for cells to develop, reconnect, and exert their therapeutic effect (around 40 weeks for human cells in the rodent brain). In such a context, brain imaging techniques that allow noninvasive longitudinal evaluation/characterization of the grafted cells in the living animals have the potential to speed-up the evaluation of candidate cell lines. This chapter describes an assemblage of imaging methods that over the years proved useful in preclinical and clinical applications for cell therapy, providing in a noninvasive way unique insights on graft cellular composition, phenotypic differentiation, as well as signs of hyperproliferation and/or immunological rejection and inflammation.


Assuntos
Doença de Huntington/terapia , Neuroimagem , Doença de Parkinson/terapia , Células-Tronco Pluripotentes/citologia , Transplante de Células-Tronco , Animais , Diferenciação Celular , Humanos
4.
Cell Stem Cell ; 15(5): 653-65, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25517469

RESUMO

Considerable progress has been made in generating fully functional and transplantable dopamine neurons from human embryonic stem cells (hESCs). Before these cells can be used for cell replacement therapy in Parkinson's disease (PD), it is important to verify their functional properties and efficacy in animal models. Here we provide a comprehensive preclinical assessment of hESC-derived midbrain dopamine neurons in a rat model of PD. We show long-term survival and functionality using clinically relevant MRI and PET imaging techniques and demonstrate efficacy in restoration of motor function with a potency comparable to that seen with human fetal dopamine neurons. Furthermore, we show that hESC-derived dopamine neurons can project sufficiently long distances for use in humans, fully regenerate midbrain-to-forebrain projections, and innervate correct target structures. This provides strong preclinical support for clinical translation of hESC-derived dopamine neurons using approaches similar to those established with fetal cells for the treatment of Parkinson's disease.


Assuntos
Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/transplante , Células-Tronco Embrionárias/citologia , Feto/citologia , Doença de Parkinson/terapia , Animais , Axônios/metabolismo , Sobrevivência Celular , Modelos Animais de Doenças , Humanos , Masculino , Mesencéfalo/embriologia , Neostriado/patologia , Fatores de Transcrição Otx/metabolismo , Doença de Parkinson/patologia , Ratos Nus , Substância Negra/patologia , Transmissão Sináptica , Fatores de Tempo
5.
Hum Mol Genet ; 22(19): 3869-82, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23720495

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder caused by an abnormal expansion of a CAG repeat encoding a polyglutamine tract in the huntingtin (Htt) protein. The mutation leads to neuronal death through mechanisms which are still unknown. One hypothesis is that mitochondrial defects may play a key role. In support of this, the activity of mitochondrial complex II (C-II) is preferentially reduced in the striatum of HD patients. Here, we studied C-II expression in different genetic models of HD expressing N-terminal fragments of mutant Htt (mHtt). Western blot analysis showed that the expression of the 30 kDa Iron-Sulfur (Ip) subunit of C-II was significantly reduced in the striatum of the R6/1 transgenic mice, while the levels of the FAD containing catalytic 70 kDa subunit (Fp) were not significantly changed. Blue native gel analysis showed that the assembly of C-II in mitochondria was altered early in N171-82Q transgenic mice. Early loco-regional reduction in C-II activity and Ip protein expression was also demonstrated in a rat model of HD using intrastriatal injection of lentiviral vectors encoding mHtt. Infection of the rat striatum with a lentiviral vector coding the C-II Ip or Fp subunits induced a significant overexpression of these proteins that led to significant neuroprotection of striatal neurons against mHtt neurotoxicity. These results obtained in vivo support the hypothesis that structural and functional alterations of C-II induced by mHtt may play a critical role in the degeneration of striatal neurons in HD and that mitochondrial-targeted therapies may be useful in its treatment.


Assuntos
Corpo Estriado/metabolismo , Complexo II de Transporte de Elétrons/metabolismo , Doença de Huntington/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Células Cultivadas , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Complexo II de Transporte de Elétrons/genética , Feminino , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Mitocôndrias/genética , Proteínas Mutantes/metabolismo , Mutação , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley
6.
PLoS One ; 4(2): e4637, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19247483

RESUMO

Aging likely plays a role in neurodegenerative disorders. In Huntington's disease (HD), a disorder caused by an abnormal expansion of a polyglutamine tract in the protein huntingtin (Htt), the role of aging is unclear. For a given tract length, the probability of disease onset increases with age. There are mainly two hypotheses that could explain adult onset in HD: Either mutant Htt progressively produces cumulative defects over time or "normal" aging renders neurons more vulnerable to mutant Htt toxicity. In the present study, we directly explored whether aging affected the toxicity of mutant Htt in vivo. We studied the impact of aging on the effects produced by overexpression of an N-terminal fragment of mutant Htt, of wild-type Htt or of a beta-Galactosidase (beta-Gal) reporter gene in the rat striatum. Stereotaxic injections of lentiviral vectors were performed simultaneously in young (3 week) and old (15 month) rats. Histological evaluation at different time points after infection demonstrated that the expression of mutant Htt led to pathological changes that were more severe in old rats, including an increase in the number of small Htt-containing aggregates in the neuropil, a greater loss of DARPP-32 immunoreactivity and striatal neurons as assessed by unbiased stereological counts.The present results support the hypothesis that "normal" aging is involved in HD pathogenesis, and suggest that age-related cellular defects might constitute potential therapeutic targets for HD.


Assuntos
Envelhecimento/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/fisiologia , Animais , Corpo Estriado/enzimologia , Corpo Estriado/patologia , Fosfoproteína 32 Regulada por cAMP e Dopamina/genética , Proteína Huntingtina , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Ratos , beta-Galactosidase/genética
7.
Neurobiol Aging ; 30(12): 2021-30, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18394756

RESUMO

Reduced expression of IGF-1R increases lifespan and resistance to oxidative stress in the mouse, raising the possibility that this also confers relative protection against the pro-parkinsonian neurotoxin MPTP, known to involve an oxidative stress component. We used heterozygous IGF-1R(+/-) mice and challenged them with MPTP. Interestingly, MPTP induced more severe lesions of dopaminergic neurons of the substantia nigra, in IGF-1R(+/-) mice than in wild-type animals. Using electron spin resonance, we found that free radicals were decreased in IGF-1R(+/-) mice in comparison with controls, both before and after MPTP exposure, suggesting that the increased vulnerability of dopamine neurons is not caused by oxidative stress. Importantly, we showed that IGF-1R(+/-) mice display a dramatically increased neuro-inflammatory response to MPTP that may ground the observed increase in neuronal death. Microarray analysis revealed that oxidative stress-associated genes, but also several anti-inflammatory signaling pathways were downregulated under control conditions in IGF-1R(+/-) mice compared to WT. Collectively, these data indicate that IGF signaling can reduce neuro-inflammation dependent sensitivity of neurons to MPTP.


Assuntos
Intoxicação por MPTP/fisiopatologia , Neuroimunomodulação/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Receptor IGF Tipo 1/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Dopamina/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Radicais Livres/metabolismo , Intoxicação por MPTP/genética , Intoxicação por MPTP/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Estresse Oxidativo/fisiologia , Receptor IGF Tipo 1/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Substância Negra/efeitos dos fármacos , Substância Negra/imunologia , Substância Negra/fisiologia
8.
Physiol Genomics ; 33(2): 170-9, 2008 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-18252803

RESUMO

Using serial analysis of gene expression, we collected quantitative transcriptome data in 11 regions of the adult wild-type mouse brain: the orbital, prelimbic, cingulate, motor, somatosensory, and entorhinal cortices, the caudate-putamen, the nucleus accumbens, the thalamus, the substantia nigra, and the ventral tegmental area. With >1.2 million cDNA tags sequenced, this database is a powerful resource to explore brain functions and disorders. As an illustration, we performed interregional comparisons and found 315 differential transcripts. Most of them are poorly characterized and 20% lack functional annotation. For 78 differential transcripts, we provide independent expression level measurements in mouse brain regions by real-time quantitative RT-PCR. We also show examples where we used in situ hybridization to achieve infrastructural resolution. For 30 transcripts, we next demonstrated that regional enrichment is conserved in the human brain. We then quantified the expression levels of region-enriched transcripts in the R6/2 mouse model of Huntington disease and the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson disease and observed significant alterations in the striatum, cerebral cortex, thalamus and substantia nigra of R6/2 mice and in the striatum of MPTP-treated mice. These results show that the gene expression data provided here for the mouse brain can be used to explore pathophysiological models and disclose transcripts differentially expressed in human brain regions.


Assuntos
Encéfalo/metabolismo , Sequência Conservada/genética , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Doenças Neurodegenerativas/genética , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Bases de Dados Genéticas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
9.
Hum Mol Genet ; 17(10): 1446-56, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18267960

RESUMO

In neurodegenerative disorders associated with primary or secondary mitochondrial defects such as Huntington's disease (HD), cells of the striatum are particularly vulnerable to cell death, although the mechanisms by which this cell death is induced are unclear. Dopamine, found in high concentrations in the striatum, may play a role in striatal cell death. We show that in primary striatal cultures, dopamine increases the toxicity of an N-terminal fragment of mutated huntingtin (Htt-171-82Q). Mitochondrial complex II protein (mCII) levels are reduced in HD striatum, indicating that this protein may be important for dopamine-mediated striatal cell death. We found that dopamine enhances the toxicity of the selective mCII inhibitor, 3-nitropropionic acid. We also demonstrated that dopamine doses that are insufficient to produce cell loss regulate mCII expression at the mRNA, protein and catalytic activity level. We also show that dopamine-induced down-regulation of mCII levels can be blocked by several dopamine D2 receptor antagonists. Sustained overexpression of mCII subunits using lentiviral vectors abrogated the effects of dopamine, both by high dopamine concentrations alone and neuronal death induced by low dopamine concentrations together with Htt-171-82Q. This novel pathway links dopamine signaling and regulation of mCII activity and could play a key role in oxidative energy metabolism and explain the vulnerability of the striatum in neurodegenerative diseases.


Assuntos
Corpo Estriado/efeitos dos fármacos , Dopamina/farmacologia , Complexo II de Transporte de Elétrons/metabolismo , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Proteínas Nucleares/genética , Animais , Técnicas de Cultura de Células , Dopaminérgicos/farmacologia , Antagonistas dos Receptores de Dopamina D2 , Regulação para Baixo/efeitos dos fármacos , Complexo II de Transporte de Elétrons/antagonistas & inibidores , Expressão Gênica/efeitos dos fármacos , Proteína Huntingtina , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Subunidades Proteicas/metabolismo , Ratos , Receptores de Dopamina D2/metabolismo
10.
Mov Disord ; 20 Suppl 12: S57-63, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16092092

RESUMO

Multiple system atrophy (MSA) is a sporadic adult-onset neurodegenerative disorder of unknown etiology clinically characterized by a combination of parkinsonian, pyramidal, and cerebellar signs. Levodopa-unresponsive parkinsonism is present in 80% of MSA cases, and this dominant clinical presentation (MSA-P) is associated with a combined degeneration of the substantia nigra pars compacta and the striatum in anatomically related areas. The limited knowledge of the pathophysiology of MSA and the lack of therapeutic strategies prompted the development of lesion models reproducing striatonigral degeneration, the substrate of levodopa-unresponsive parkinsonism in MSA-P. This method was carried out first in rats with two different stereotaxic strategies using either two neurotoxins ("double toxin-double lesion") or a single neurotoxin ("single toxin-double lesion"). Double-lesioned rat models showed severe motor impairment compared to those with a single nigral or striatal lesion and helped to mimic different stages of the disease. Systemic models were also developed in mice and primates using the nigral toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and the striatal toxin 3-nitropropionic (3-NP). In mice, although MPTP reduced the subsequent sensitivity to 3-NP in a sequential lesion, simultaneous nigral and striatal insults were shown to exacerbate striatal damage. MPTP-treated monkeys displayed a significant worsening of parkinsonism and a loss of levodopa-responsiveness after the appearance of hindlimb dystonia and striatal lesion formation induced by subsequent 3-NP intoxication. The different species and intoxication paradigms used will be useful to investigate functional changes in substantia nigra and striatum and to define neuroprotective, neurorestorative, or symptomatic therapeutic strategies.


Assuntos
Atrofia de Múltiplos Sistemas/patologia , Animais , Antiparkinsonianos/uso terapêutico , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Levodopa/uso terapêutico , Intoxicação por MPTP/metabolismo , Macaca fascicularis , Camundongos , Atrofia de Múltiplos Sistemas/genética , Atrofia de Múltiplos Sistemas/metabolismo , Oxidopamina/metabolismo , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/patologia , Mutação Puntual/genética , Ratos , Substância Negra/metabolismo , Substância Negra/patologia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
11.
J Neurosci Res ; 78(1): 87-91, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15372496

RESUMO

Previous studies in rodent models of neurodegenerative disorders have demonstrated that minocycline exerts neuroprotective effects unrelated to its antimicrobial action. The purpose of the present study was to analyze whether minocycline exhibits neuroprotective activity in a rat model of striatonigral degeneration (SND), the core pathology underlying levodopa-unresponsive parkinsonism associated with multiple system atrophy (MSA). We observed no significant effect of minocycline on locomotor impairment in double-lesioned SND rats. Minocycline significantly suppressed astroglial and microglial activation (P < 0.01); however, 3'5'-monophosphate-regulated phosphoprotein (DARPP 32) immunohistochemistry revealed no significant differences in striatal lesion volume of minocycline-treated versus untreated control SND rats. Furthermore, there was no protection of nigral dopaminergic neurons in the double-lesion model. We conclude that despite its astrocytic and microglial suppression, minocycline failed to attenuate lesion-induced neuronal damage in the SND rat model.


Assuntos
Modelos Animais de Doenças , Neuroglia/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Degeneração Estriatonigral/metabolismo , Animais , Masculino , Minociclina/farmacologia , Minociclina/uso terapêutico , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Wistar , Degeneração Estriatonigral/prevenção & controle
12.
Exp Neurol ; 189(1): 1-4, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15296829

RESUMO

Initial studies conducted on the neuroprotective effects of minocycline, a second-generation tetracycline, in experimental models of neurodegeneration gave promising results. However, more recently, minocycline has clearly been shown to have variable and even contradictory (beneficial or detrimental) effects in different species and models of neurological disorders, and its "neuroprotective" mechanisms remain to be clarified. Although its anti-inflammatory properties are likely to contribute to its neuroprotective effects observed in several animal models, a body of recent evidence indicates that our community should proceed with caution in the clinical use of minocycline for central nervous system disorders.


Assuntos
Doenças do Sistema Nervoso Central/prevenção & controle , Minociclina/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Humanos
13.
Eur J Neurosci ; 19(12): 3266-76, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15217383

RESUMO

Minocycline has been shown to exert anti-inflammatory effects underlying its putative neuroprotective properties in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease and in the R6/2 mouse model of Huntington's disease (HD). However, contradictory results have recently been reported. We report deleterious effects of minocycline in two phenotypic (toxic) models of Parkinson's disease and HD in monkey and mouse. Of seven MPTP-intoxicated female cynomolgus monkeys (0.2 mg/kg, i.v. until day 15), three received minocycline (200 mg b.i.d.). While placebo-MPTP-treated animals displayed mild parkinsonism at day 15, the minocycline/MPTP-treated animals tended to be more affected (P = 0.057) and showed a greater loss of putaminal dopaminergic nerve endings (P < 0.0001). In the 3-nitropropionic acid (3-NP) mouse model of HD, minocycline (45 mg/kg i.p.) was administered 30 min before each i.p. injection of 3-NP (b.i.d., cumulated dose, 360 mg/kg in 5 days). Mice receiving minocycline exhibited a worsening of the mean motor score with a slower recovery slope, more impaired general activity and significantly deteriorated performances on the rotarod, pole test and beam-traversing tasks. The histopathological outcome demonstrated that minocycline-treated mice presented significantly more severe neuronal cell loss in the dorsal striatum. The effect of minocycline vs. 3-NP was also investigated on hippocampal and cortical cell cultures. minocycline blocked 3-NP-induced neurotoxicity at certain doses (1 mm cortical neurons) but not at higher doses (10 mm). Thus, minocycline may have variable and even deleterious effects in different species and models according to the mode of administration and dose.


Assuntos
Corpo Estriado/patologia , Doença de Huntington/tratamento farmacológico , Minociclina/efeitos adversos , Fármacos Neuroprotetores/efeitos adversos , Transtornos Parkinsonianos/tratamento farmacológico , Animais , Células Cultivadas , Convulsivantes/toxicidade , Corpo Estriado/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Doença de Huntington/induzido quimicamente , Imuno-Histoquímica , Macaca fascicularis , Masculino , Camundongos , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Nitrocompostos , Propionatos/toxicidade
14.
Neurobiol Dis ; 15(3): 667-75, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15056475

RESUMO

Age of onset of Huntington's disease (HD) statistically correlates with the length of expanded CAG repeats in the IT15 gene. However, other factors such as polymorphism in the 3' untranslated region of the GluR6 kainate receptor gene subunit may contribute to variability in the age at onset. To investigate this issue, we studied the motor disorder and related striatal damage induced by 3-nitropropionic acid (3-NP) subacute administration in GluR6 knockout mice (GluR6(-/-)) as compared to wild-type mice. In two different age groups (6 months and 1 year), we observed that GluR6(-/-) mice did not display more motor impairment nor more striatal histopathological damage than GluR6(+/+) mice, although 1-year-old GluR6(-/-) mice displayed reduced activity parameters either at baseline or after 3-NP administration compared to GluR6(+/+). In both age groups, GluR6(-/-) mice died earlier and displayed earlier motor symptoms during 3-NP-induced metabolic compromise, suggesting that GluR6-containing kainate receptors may be implicated during neurodegeneration, such as in HD, rather than in the final outcome.


Assuntos
Corpo Estriado/metabolismo , Doença de Huntington/metabolismo , Receptores de Ácido Caínico/metabolismo , Idade de Início , Animais , Comportamento Animal , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Genótipo , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Nitrocompostos , Propionatos/farmacologia , Receptores de Ácido Caínico/genética , Succinato Desidrogenase/metabolismo , Receptor de GluK2 Cainato
16.
Eur J Neurosci ; 15(12): 2053-6, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12099912

RESUMO

Evidence suggests that dopamine is involved in the modulation of striatal excitotoxic processes. To further investigate this issue, we studied the effects of systemic 'low-dose' (total dose, 340 mg/kg in 7 days) 3-nitropropionic acid (3-NP) intoxication in dopamine transporter knock-out mice (DAT-/-) compared to wildtype (DAT+/+) mice. Systemic 'low-dose' 3-NP induced a significant impairment in a rotarod task only in DAT-/- mice. Histopathology also demonstrated a significant reduction of the striatal volume (-7%, P < 0.05), neuronal density (-12.5%, P < 0.001) and absolute number estimates of striatal neurons (-11.5%, P < 0.001) in DAT-/- compared to DAT+/+ mice, with increased glial activation, independent of the degree of succinate dehydrogenase inhibition. These findings strengthen the hypothesis for dopamine modulation of excitotoxicity within the nigrostriatal system.


Assuntos
Doenças dos Gânglios da Base/metabolismo , Dopamina/metabolismo , Tolerância a Medicamentos/genética , Glicoproteínas de Membrana , Proteínas de Membrana Transportadoras/deficiência , Neostriado/metabolismo , Proteínas do Tecido Nervoso , Vias Neurais/metabolismo , Substância Negra/metabolismo , Animais , Doenças dos Gânglios da Base/induzido quimicamente , Doenças dos Gânglios da Base/fisiopatologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Convulsivantes/farmacologia , Proteínas da Membrana Plasmática de Transporte de Dopamina , Relação Dose-Resposta a Droga , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Neostriado/lesões , Neostriado/fisiopatologia , Degeneração Neural/induzido quimicamente , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Vias Neurais/lesões , Vias Neurais/fisiopatologia , Neurotoxinas/farmacologia , Nitrocompostos , Equilíbrio Postural/efeitos dos fármacos , Equilíbrio Postural/fisiologia , Propionatos/farmacologia , Substância Negra/lesões , Substância Negra/fisiopatologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
17.
J Neurosci Methods ; 113(2): 123-30, 2002 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-11772434

RESUMO

Reduced stride length characterizes Parkinsonian gait. We aimed to demonstrate that it could be measured simply and reliably in mice by pawprints and used as an index of basal ganglia dysfunction. In C57BL/6 mice, stride length measurements proved to be consistent across measurements and experimenters. It was slightly lower in the hindlimbs and was correlated to femur size and animal velocity. Dopamine depletion by reserpine and striatal dopamine receptor blockade by haloperidol resulted in reduced mean stride length in four limbs. Significant forelimb/hindlimb difference was also observed both in mice with 3-nitropropionic acid (3-NP) induced striatal lesions and in those with MPTP-induced nigral cell loss. Reduction of hindlimb stride length was correlated significantly with the magnitude of cell loss, either in the substantia nigra or in the lateral mid-striatum. Stride length is, therefore, a simple method to obtain an index of motor disorders due to basal ganglia dysfunction in mice.


Assuntos
Corpo Estriado/fisiopatologia , Marcha/fisiologia , Transtornos Parkinsonianos/fisiopatologia , Substância Negra/fisiopatologia , Animais , Antipsicóticos/farmacologia , Benserazida/farmacologia , Convulsivantes , Dopaminérgicos/farmacologia , Antagonistas de Dopamina/farmacologia , Feminino , Marcha/efeitos dos fármacos , Haloperidol/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nitrocompostos , Propionatos , Reserpina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA