Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Med Chem ; 57(20): 8293-306, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25211466

ABSTRACT

This paper reports the design and synthesis of C-glycoside mimetics (d-glycero-d-talo- and d-glycero-d-galactopyranose analogues), a subset of the recently published phostines, belonging to the [1,2]oxaphosphinane core. Eighteen new compounds were tested against 11 cancer cell types belonging to six categories of tumor tissues and three different species. The hit compound 5.3d inhibited invasion and migration of both GBM stem cells (Gli7 and Gli4) and GBM cancer cell lines (C6, SNB75) on fibronectin, vitronectin, and laminin. Ki values for Gli7 and Gli4 migration inhibition on fibronectin were 16 and 31 nM respectively. Ki values for invasion inhibition in a 3D system were 46 nM for Gli7 and 290 nM for Gli4. These activities were associated with an antiproliferative effect on Gli4 (EC50 = 5.20 µM) and Gli7 (EC50 = 2.33 µM). In conclusion, the heptopyranose mimetic 5.3d, devoid of toxicity on astrocyte and cortical neuron cultures at concentrations below 100 µM, opens new therapeutic perspectives against glioblastoma.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Glioma/drug therapy , Monosaccharides/chemistry , Neoplastic Stem Cells/drug effects , Animals , Astrocytes/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemistry Techniques, Synthetic , Drug Screening Assays, Antitumor , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioma/pathology , Glycosides , Humans , Mice , Molecular Mimicry , Molecular Structure , Neoplastic Stem Cells/pathology , Neurons/drug effects , Rats
2.
PLoS One ; 7(9): e44787, 2012.
Article in English | MEDLINE | ID: mdl-22970307

ABSTRACT

Ovarian cancer is the gynecological cancer exhibiting the highest morbidity and improvement of treatments is still required. Previous studies have shown that Estrogen-receptor beta (ERß) levels decreased along with ovarian carcinogenesis. Here, we present evidence that reintroduction of ERß in BG-1 epithelial ovarian cancer cells, which express ERα, leads in vitro to a decrease of basal and estradiol-promoted cell proliferation. ERß reduced the frequency of cells in S phase and increased the one of cells in G2/M phase. At the molecular level, we found that ERß downregulated total retinoblastoma (Rb), phosphorylated Rb and phospho-AKT cellular content as well as cyclins D1 and A2. In addition, ERß had a direct effect on ERα, by strongly inhibiting its expression and activity, which could explain part of the anti-proliferative action of ERß. By developing a novel preclinical model of ovarian cancer based on a luminescent orthotopic xenograft in athymic Nude mice, we further revealed that ERß expression reduces tumor growth and the presence of tumor cells in sites of metastasis, hence resulting in improved survival of mice. Altogether, these findings unveil a potential tumor-suppressor role of ERß in ovarian carcinogenesis, which could be of potential clinical relevance for the selection of the most appropriate treatment for patients.


Subject(s)
Estrogen Receptor beta/physiology , Genes, Tumor Suppressor , Neoplasms, Glandular and Epithelial/physiopathology , Ovarian Neoplasms/physiopathology , Cell Proliferation , Estrogen Receptor beta/genetics , Female , Humans , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphorylation , Retinoblastoma Protein/metabolism , Transcription, Genetic
3.
J Med Chem ; 55(5): 2196-211, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22268526

ABSTRACT

This paper reports the design and the synthesis of a new family of compounds, the phostines, belonging to the [1,2]oxaphosphinane family. Twenty-six compounds have been screened for their antiproliferative activity against a large panel of NCI cancer cell lines. Because of its easy synthesis and low EC(50) value (500 nM against the C6 rat glioma cell line), compound 3.1a was selected for further biological study. Moreover, the specific biological effect of 3.1a on the glioblastoma phylogenetic cluster from the NCI is dependent on its stereochemistry. Within that cluster, 3.1a has a higher antiproliferative activity than Temozolomide and is more potent than paclitaxel for the SF295 and SNB75 cell lines. In constrast with paclitaxel and vincristine, 3.1a is devoid of astrocyte toxicity. The original activity spectrum of 3.1a on the NCI cancer cell line panel allows the development of this family for use in association with existing drugs, opening new therapeutic perspectives.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cyclic P-Oxides/chemical synthesis , Organophosphonates/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Astrocytes/cytology , Astrocytes/drug effects , Brain Neoplasms/drug therapy , Cell Count , Cell Line, Tumor , Cell Survival/drug effects , Cyclic P-Oxides/chemistry , Cyclic P-Oxides/pharmacology , Drug Screening Assays, Antitumor , Glioblastoma/drug therapy , Humans , Organophosphonates/chemistry , Organophosphonates/pharmacology , Phosphorous Acids , Rats , Stereoisomerism , Structure-Activity Relationship
4.
Neurobiol Aging ; 32(11): 2100-2, 2011 Nov.
Article in English | MEDLINE | ID: mdl-20018410

ABSTRACT

Supporting the hypothesis that proteasome dysfunction is involved in Parkinson's disease (PD), McNaught et al. (2004) reported that the systemic administration of the proteasome inhibitor Z-Ile-Glu(OtBu)-Ala-Leu-aldehyde (PSI) in rats led to the degeneration of the nigrostriatal pathway. However, several groups could not reproduce this finding. We herein attempted to improve the reliability of the PSI model by chronically delivering the inhibitor using osmotic minipumps in aged mice. We also tested whether PSI co-administered with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) could act synergistically to induce toxicity. We found that PSI produced a significant reduction in locomotor activity that was mildly exacerbated by MPTP. However, PSI alone produced no sign of degeneration of the nigrostriatal dopaminergic pathway and did not exacerbate MPTP toxicity. To conclude, PSI administration does not provide a reliable phenotypic model of PD.


Subject(s)
Corpus Striatum/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Hypokinesia/chemically induced , Motor Activity/drug effects , Oligopeptides/pharmacology , Substantia Nigra/drug effects , Animals , Mice , Neurons/drug effects
5.
Neurobiol Dis ; 40(1): 311-24, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20600911

ABSTRACT

In neurodegenerative disorders caused by polyglutamine (polyQ) expansion, polyQ toxicity is thought to trigger a linear cascade of successive degenerative events leading to neuronal death. To understand how neurons cope with polyQ toxicity, we studied a Spinocerebellar ataxia 7 (SCA7) mouse which expresses polyQ-expanded ATXN7 only in rod photoreceptors. We show that in response to polyQ toxicity, SCA7 rods go through a range of radically different cell fates, including apoptotic and non-apoptotic cell death, cell migration, morphological transformation into a round cell or, most remarkably, cell division. The temporal profile of retinal remodeling indicates that some degenerative pathways are triggered early in the disease but decline later on, while others worsen progressively. Retinal remodeling results in a relative maintenance of photoreceptor population, but does not preserve the retinal function. Rod responses to proteotoxicity correlate with the nature, level and ratio of mutant ATXN7 species. The multifaceted response of neurons to polyQ toxicity is an important concept for the design of therapeutic strategies.


Subject(s)
Nerve Tissue Proteins/metabolism , Peptides/toxicity , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/pathology , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology , Animals , Apoptosis Regulatory Proteins/genetics , Ataxin-7 , Cell Death/physiology , Cell Movement/genetics , Cell Shape/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/toxicity , Retinal Degeneration/etiology , Retinal Rod Photoreceptor Cells/metabolism , Spinocerebellar Ataxias/complications
6.
Neurochem Int ; 56(2): 208-12, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19852992

ABSTRACT

Promethazine (PMZ), an FDA-approved antihistaminergic drug, was identified as a potentially neuroprotective compound in a NINDS screening program. It was shown to protect against ischemia in mice, to delay disease onset in a mouse model of amyotrophic lateral sclerosis and to inhibit Ca(2+)-induced mitochondrial permeability transition in rat liver mitochondria. We investigated whether PMZ could protect against the neurotoxic effects induced by 3-nitropropionic acid (3-NP), an inhibitor of the succinate dehydrogenase, used to model Huntington's disease (HD) in rats. Lewis rats receiving chronic subcutaneous infusion of 3-NP were treated with PMZ. The findings indicate that chronic PMZ treatment significantly reduced 3-NP-induced striatal lesion volume, loss of GABAergic neurons and number of apoptotic cells in the striatum. PMZ showed a strong neuroprotective effect against 3-NP toxicity in vivo.


Subject(s)
Amyotrophic Lateral Sclerosis/prevention & control , Brain Ischemia/prevention & control , Huntington Disease/prevention & control , Neuroprotective Agents/pharmacology , Nitro Compounds/toxicity , Promethazine/pharmacology , Propionates/toxicity , Amyotrophic Lateral Sclerosis/chemically induced , Animals , Brain Ischemia/chemically induced , Disease Models, Animal , Huntington Disease/chemically induced , Male , Mice , Rats , Rats, Inbred Lew , Succinate Dehydrogenase/metabolism
7.
J Neurosci ; 26(22): 5978-89, 2006 May 31.
Article in English | MEDLINE | ID: mdl-16738240

ABSTRACT

To study the functional role of activated astrocytes in glutamate homeostasis in vivo, we used a model of sustained astrocytic activation in the rat striatum through lentiviral-mediated gene delivery of ciliary neurotrophic factor (CNTF). CNTF-activated astrocytes were hypertrophic, expressed immature intermediate filament proteins and highly glycosylated forms of their glutamate transporters GLAST and GLT-1. CNTF overexpression produced a redistribution of GLAST and GLT-1 into raft functional membrane microdomains, which are important for glutamate uptake. In contrast, CNTF had no detectable effect on the expression of a number of neuronal proteins and on the spontaneous glutamatergic transmission recorded from striatal medium spiny neurons. These results were replicated in vitro by application of recombinant CNTF on a mixed neuron/astrocyte striatal culture. Using microdialysis in the rat striatum, we found that the accumulation of extracellular glutamate induced by quinolinate (QA) was reduced threefold with CNTF. In line with this result, CNTF significantly increased QA-induced [(18)F]-fluoro-2-deoxyglucose uptake, an indirect index of glutamate uptake by astrocytes. Together, these data demonstrate that CNTF activation of astrocytes in vivo is associated with marked phenotypic and molecular changes leading to a better handling of increased levels of extracellular glutamate. Activated astrocytes may therefore be important prosurvival agents in pathological conditions involving defects in glutamate homeostasis.


Subject(s)
Astrocytes/physiology , Ciliary Neurotrophic Factor/pharmacology , Corpus Striatum/physiology , Excitatory Amino Acid Transporter 1/physiology , Excitatory Amino Acid Transporter 2/physiology , Glutamic Acid/physiology , Membrane Microdomains/physiology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Ciliary Neurotrophic Factor/genetics , Coculture Techniques , Corpus Striatum/drug effects , Excitatory Amino Acid Transporter 1/drug effects , Excitatory Amino Acid Transporter 2/drug effects , Genetic Vectors , Humans , Lentivirus/genetics , Male , Membrane Microdomains/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Rats , Rats, Inbred Lew
8.
FASEB J ; 20(7): 1021-3, 2006 May.
Article in English | MEDLINE | ID: mdl-16571773

ABSTRACT

According to the "indirect" excitotoxicity hypothesis, mitochondrial defects increase Ca2+ entry into neurons by rendering NMDA-R hypersensitive to glutamate. We tested this hypothesis by investigating in the rat striatum and cultured striatal cells how partial mitochondrial complex II inhibition produced by 3-nitropropionic acid (3NP) modifies the toxicity of the NMDA-R agonist quinolinate (QA). We showed that nontoxic 3NP treatment, leading to partial inhibition of complex II activity, greatly exacerbated striatal degeneration produced by slightly toxic QA treatment through an "all-or-nothing" process. The potentiation of QA-induced cell death by 3NP was associated with increased calpain activity and massive calpain-mediated cleavage of several postsynaptic proteins, suggesting major neuronal Ca2+ deregulation in the striatum. However, Ca2+ anomalies probably do not result from NMDA-R hypersensitivity. Indeed, brain imaging experiments using [(18)F]fluorodeoxyglucose indirectly showed that 3NP did not increase QA-induced ionic perturbations at the striatal glutamatergic synapses in vivo. Consistent with this, the exacerbation of QA toxicity by 3NP was not related to an increase in the QA-induced entry of 45Ca2+ into striatal neurons. The present results demonstrate that the potentiation of NMDA-R-mediated excitotoxicity by mitochondrial defects involves primarily intracellular Ca2+ deregulation, in the absence of NMDA-R hypersensitivity.


Subject(s)
Calcium Signaling/physiology , Corpus Striatum/metabolism , Mitochondria/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calpain/metabolism , Cells, Cultured , Corpus Striatum/drug effects , Corpus Striatum/pathology , Male , Mitochondria/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nitro Compounds/pharmacology , Propionates/pharmacology , Quinolinic Acid/adverse effects , Rats , Rats, Inbred Lew
9.
J Neurochem ; 95(6): 1521-40, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16300642

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by a mutation in the gene encoding Huntingtin. The mechanisms underlying the preferential degeneration of the striatum, the most striking neuropathological change in HD, are unknown. Of those probably involved, mitochondrial defects might play an important role. The behavioural and anatomical similarities found between HD and models using the mitochondrial toxin 3-nitropropionic acid (3NP) in rats and primates support this hypothesis. Here, we discuss the recently identified mechanisms of 3NP-induced striatal degeneration. Two types of important factor have been identified. The first are the 'executioner' components that have direct roles in cell death, such as c-Jun N-terminal kinase and Ca2+-activated protease calpains. The second are 'environmental' factors, such as glutamate, dopamine and adenosine, which modulate the striatal degeneration induced by 3NP. Interestingly, these recent studies support the hypothesis that 3NP and mutated Huntingtin have certain mechanisms of toxicity in common, suggesting that the use of 3NP might give new insights into the pathogenesis of HD. Research on 3NP provides additional proof that the neurochemical environment of a given neurone can determine its preferential vulnerability in neurodegenerative diseases.


Subject(s)
Huntington Disease/physiopathology , Mitochondria/drug effects , Neostriatum/physiopathology , Neurotoxins , Nitro Compounds , Propionates , Animals , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Mitochondria/metabolism , Neostriatum/pathology , Nerve Degeneration/pathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Phenotype
10.
Neuropharmacology ; 49(5): 695-702, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15998526

ABSTRACT

The contribution of calpains and caspases to cell death has been widely studied using pharmacological inhibitors. Among them, the caspase inhibitor N-benzyloxycarbonyl-valyl-alanyl-aspartyl-fluoromethylketone (zVAD) has been used as a specific caspase inhibitor in nearly 1000 published studies. However, several studies showed that zVAD also behaves as a calpain inhibitor in peripheral cells. The effects of zVAD as a calpain inhibitor have never been assessed in neurodegeneration models. We examined here whether zVAD could reduce neurodegeneration in Huntington's disease models using the mitochondrial inhibitor 3-nitropropionic acid (3NP). In these models, 3NP toxicity has been shown to require calpain activation. In rats, intra-cerebro-ventricular infusion of zVAD significantly reduced 3NP-induced striatal degeneration, and decreased the 3NP-induced activation of calpain and calpain-dependent cleavage of fodrin. zVAD (100 microM) also blocked 3NP-induced death of cultured striatal neurons. In vitro, zVAD inhibited purified mu-calpain with high affinity (IC50=10 nM). The present data demonstrate that zVAD protects neurons against 3NP through calpain inhibition. This suggests that, in certain models of neuronal death where zVAD showed protective effects, caspases but also calpains may be involved.


Subject(s)
Calpain/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Propionates/antagonists & inhibitors , Propionates/toxicity , Protease Inhibitors , Animals , Calpain/metabolism , Carrier Proteins/metabolism , Cell Death/drug effects , Huntington Disease/pathology , Immunohistochemistry , Injections, Intraventricular , Male , Microfilament Proteins/metabolism , Neostriatum/cytology , Neostriatum/drug effects , Nerve Degeneration/pathology , Neuroprotective Agents/administration & dosage , Nitro Compounds , Oligopeptides/administration & dosage , Rats , Rats, Inbred Lew , Rats, Wistar
11.
Neurobiol Dis ; 18(1): 206-17, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15649711

ABSTRACT

Minocycline has been shown to be neuroprotective in various models of neurodegenerative diseases. However, its potential in Huntington's disease (HD) models characterized by calpain-dependent degeneration and inflammation has not been investigated. Here, we have tested minocycline in phenotypic models of HD using 3-nitropropionic acid (3NP) intoxication and quinolinic acid (QA) injections. In the 3NP rat model, where the development of striatal lesions involves calpain, we found that minocycline was not protective, although it attenuated the development of inflammation induced after the onset of striatal degeneration. The lack of minocycline activity on calpain-dependent cell death was also confirmed in vitro using primary striatal cells. Conversely, we found that minocycline reduced lesions and inflammation induced by QA. In cultured cells, minocycline protected against mutated huntingtin and staurosporine, stimulations known to promote caspase-dependent cell death. Altogether, these data suggested that, in HD, minocycline may counteract the development of caspase-dependent neurodegeneration, inflammation, but not calpain-dependent neuronal death.


Subject(s)
Huntington Disease/drug therapy , Minocycline/pharmacology , Nerve Degeneration/drug therapy , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , Animals , Calpain/drug effects , Calpain/metabolism , Caspases/drug effects , Caspases/metabolism , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Corpus Striatum/drug effects , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalitis/drug therapy , Encephalitis/physiopathology , Encephalitis/prevention & control , Glutamic Acid/metabolism , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Male , Minocycline/therapeutic use , Nerve Degeneration/pathology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/therapeutic use , Nitro Compounds , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Phenotype , Propionates , Quinolinic Acid , Rats , Rats, Inbred Lew , Rats, Wistar , Staurosporine/antagonists & inhibitors
12.
J Biol Chem ; 278(44): 43245-53, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-12917435

ABSTRACT

The role of caspases and calpains in neurodegeneration remains unclear. In this study, we focused on these proteases in a rat model of Huntington's disease using the mitochondrial toxin 3-nitropropionic acid (3NP). Results showed that 3NP-induced death of striatal neurons was preceded by cytochrome c redistribution, transient caspase-9 processing, and activation of calpain, whereas levels of the active/processed form of caspase-3 remained low and were even reduced as compared with control animals. We evidenced here that this decrease in active caspase-3 levels could be attributed to calpain activation. Several observations supported this conclusion. 1) Pharmacological blockade of calpain in 3NP-treated rats increased the levels of endogenous processed caspase-9 and caspase-3. 2) Cell-free extracts prepared from the striatum of 3NP-treated rats degraded in vitro the p34 and p20 subunits of active recombinant caspase-9 and caspase-3, respectively. 3) This degradation of p34 and p20 could be mimicked by purified mu-calpain and was prevented by calpain inhibitors. 4) mu-Calpain produced a loss of the DEVDase (Asp-Glu-Val-Asp) activity of active caspase-3. 5) Western blot analysis and experiments with 35S-radiolabeled caspase-3 showed that mu-calpain cleaved the p20 subunit of active caspase-3 near its catalytic site. 6) mu-Calpain activity was selectively inhibited (IC50 of 100 mum) by a 12 amino acid peptide corresponding to the C terminus of p20. Our results showed that calpain can down-regulate the caspase-9/caspase-3 cell death pathway during neurodegeneration due to chronic mitochondrial defects in vivo and that this effect may involve, at least in part, direct cleavage of the caspase-3 p20 subunit.


Subject(s)
Brain/metabolism , Calpain/metabolism , Caspases/metabolism , Propionates/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Caspase 3 , Caspase 9 , Cell-Free System , Cytosol/metabolism , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Inhibitory Concentration 50 , Male , Models, Biological , Molecular Sequence Data , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Nitro Compounds , Protein Binding , Protein Structure, Tertiary , Rats , Rats, Inbred Lew , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Time Factors
13.
J Neurosci ; 23(12): 5020-30, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12832525

ABSTRACT

Striatal cell death in Huntington's Disease (HD) may involve mitochondrial defects, NMDA-mediated excitotoxicity, and activation of death effector proteases such as caspases and calpain. However, the precise contribution of mitochondrial defects in the activation of these proteases in HD is unknown. Here, we addressed this question by studying the mechanism of striatal cell death in rat models of HD using the mitochondrial complex II inhibitor 3-nitropropionic acid (3-NP). The neurotoxin was either given by intraperitoneal injections (acute model) or over 5 d by constant systemic infusion using osmotic pumps (chronic model) to produce either transient or sustained mitochondrial deficits. Caspase-9 activation preceded neurodegeneration in both cases. However, caspase-8 and caspase-3 were activated in the acute model, but not in the chronic model, showing that 3-NP does not require activation of these caspases to produce striatal degeneration. In contrast, activation of calpain was specifically detected in the striatum in both models and this was associated with a calpain-dependent cleavage of huntingtin. Finally, in the chronic model, which mimics a steady blockade of complex II activity reminiscent of HD, selective calpain inhibition prevented the abnormal calpain-dependent processing of huntingtin, reduced the size of the striatal lesions, and almost completely abolished the 3-NP-induced DNA fragmentation in striatal cells. The present results demonstrate that calpain is a predominant effector of striatal cell death associated with mitochondrial defects in vivo. This suggests that calpain may play an important role in HD pathogenesis and could be a potential therapeutic target to slow disease progression.


Subject(s)
Calpain/metabolism , Corpus Striatum/metabolism , Huntington Disease/metabolism , Propionates , Acute Disease , Animals , Calpain/antagonists & inhibitors , Caspases/metabolism , Cell Death , Chronic Disease , Corpus Striatum/drug effects , Corpus Striatum/pathology , DNA Fragmentation/drug effects , Disease Models, Animal , Drug Administration Routes , Electron Transport Complex II , Enzyme Inhibitors/pharmacology , Huntingtin Protein , Huntington Disease/chemically induced , Huntington Disease/pathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Multienzyme Complexes/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Nitro Compounds , Nuclear Proteins/metabolism , Oxidoreductases/antagonists & inhibitors , Rats , Rats, Inbred Lew , Succinate Dehydrogenase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...