Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Medicinas Complementares
Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Neurotox Res ; 36(2): 376-386, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31201732

RESUMO

Repeated methamphetamine (METH) exposure can cause severe neurotoxicity to the central nervous system, and lead to memory deficits. L-Stepholidine (L-SPD) is a structurally identified alkaloid extract of the Chinese herb Stephania intermedia, which elicits dopamine (DA) D1-type receptors partial agonistic activity and D2-type receptors antagonistic activity. In this study, we investigated the effect of L-SPD on METH-induced memory deficits in mice and its underlying mechanisms. We found that repeated exposure to METH (10 mg/kg, i.p., once per day for 7 consecutive days) impaired memory functions in the novel object recognition experiment. Pretreatment of L-SPD (10 mg/kg, i.p.) significantly improved METH-induced memory deficits in mice. Meanwhile, the protein expression of dopaminergic D2 receptors in hippocampus area was significantly increased by repeated METH exposure, while the protein expression of dopamine transporter (DAT) was significantly reduced. Additionally, the protein expression of phospho-protein kinase A (p-PKA) was significantly increased by repeated METH exposure. The hyperpolarization-activated cyclic-nucleotide-gated non-selective cation 1 (HCN1) channel, which was a key regulator of memory functions and could be regulated by p-PKA, was also significantly increased by repeated METH exposure. These changes caused by METH could be prevented by L-SPD pretreatment. Therefore, our data firstly showed that pretreatment of L-SPD exhibited the protective effect against METH-induced memory deficits, possibly through reducing METH-induced upregulation of dopaminergic pathway and HCN1 channels.


Assuntos
Berberina/análogos & derivados , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/prevenção & controle , Metanfetamina/toxicidade , Fármacos Neuroprotetores/uso terapêutico , Animais , Berberina/uso terapêutico , Dopaminérgicos/toxicidade , Agonistas de Dopamina/uso terapêutico , Antagonistas de Dopamina/uso terapêutico , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória
2.
Nutrients ; 10(9)2018 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-30131460

RESUMO

Mucuna pruriens (Mucuna) has been prescribed in Ayurveda for various brain ailments including 'kampavata' (tremors) or Parkinson's disease (PD). While Mucuna is a well-known natural source of levodopa (L-dopa), published studies suggest that other bioactive compounds may also be responsible for its anti-PD effects. To investigate this hypothesis, an L-dopa reduced (<0.1%) M. pruriens seeds extract (MPE) was prepared and evaluated for its anti-PD effects in cellular (murine BV-2 microglia and human SH-SY5Y neuroblastoma cells), Caenorhabditis elegans, and Drosophila melanogaster models. In BV-2 cells, MPE (12.5⁻50 µg/mL) reduced hydrogen peroxide-induced cytotoxicity (15.7-18.6%), decreased reactive oxygen species production (29.1-61.6%), and lowered lipopolysaccharide (LPS)-induced nitric oxide species release by 8.9⁻60%. MPE (12.5-50 µg/mL) mitigated SH-SY5Y cell apoptosis by 6.9-40.0% in a non-contact co-culture assay with cell-free supernatants from LPS-treated BV-2 cells. MPE (12.5-50 µg/mL) reduced 6-hydroxydopamine (6-OHDA)-induced cell death of SH-SY5Y cells by 11.85⁻38.5%. Furthermore, MPE (12.5-50 µg/mL) increased median (25%) and maximum survival (47.8%) of C. elegans exposed to the dopaminergic neurotoxin, methyl-4-phenylpyridinium. MPE (40 µg/mL) ameliorated dopaminergic neurotoxin (6-OHDA and rotenone) induced precipitation of innate negative geotaxis behavior of D. melanogaster by 35.3 and 32.8%, respectively. Therefore, MPE contains bioactive compounds, beyond L-dopa, which may impart neuroprotective effects against PD.


Assuntos
Levodopa/farmacologia , Microglia/efeitos dos fármacos , Mucuna/química , Fármacos Neuroprotetores/farmacologia , Extratos Vegetais/farmacologia , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Dopaminérgicos/toxicidade , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/metabolismo , Humanos , Masculino , Microglia/metabolismo , Neuroblastoma/metabolismo , Oxidopamina/toxicidade , Doença de Parkinson/tratamento farmacológico , Espécies Reativas de Oxigênio/metabolismo , Sementes/química
3.
Neurotox Res ; 24(2): 288-97, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23575992

RESUMO

Production of nitric oxide (NO) has been implicated in methamphetamine (METH)-induced dopamine (DA) neurotoxicity. The source of this NO has not been clearly delineated, but recent evidence suggests that it arises from activation of neuronal nitric oxide synthase (nNOS), which is selectively expressed in a subpopulation of striatal interneurons. Our objective was to determine whether inhibiting activation of nNOS-containing interneurons in the striatum blocks METH-induced neurotoxicity. These interneurons selectively express the neurokinin-1 (NK-1) receptor, which is activated by substance P. One particular toxin, a conjugate of substance P to the ribosome-inactivating protein saporin (SSP-SAP), selectively destroys neurons expressing the NK-1 receptor. Thus, we examined the extent to which depletion of the nNOS-containing interneurons alters production of NO and attenuates METH-induced neurotoxicity. The SSP-SAP lesions resulted in significant loss of nNOS-containing interneurons throughout striatum. Surprisingly, this marked deletion did not confer resistance to METH-induced DA neurotoxicity, even in areas devoid of nNOS-positive cells. Furthermore, these lesions did not attenuate NO production, even in areas lacking nNOS. These data suggest that nNOS-containing interneurons either are not necessary for METH-induced DA neurotoxicity or produce NO that can diffuse extensively through striatal tissue and thereby still mediate neurotoxicity.


Assuntos
Corpo Estriado/enzimologia , Dopaminérgicos/toxicidade , Interneurônios/enzimologia , Metanfetamina/toxicidade , Óxido Nítrico Sintase Tipo I/fisiologia , Animais , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Avaliação Pré-Clínica de Medicamentos/métodos , Interneurônios/efeitos dos fármacos , Interneurônios/patologia , Masculino , Ratos , Ratos Sprague-Dawley
4.
Brain Res ; 1508: 53-62, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23499562

RESUMO

The 3-O-demethylswertipunicoside (3-ODS) is extracted from Swertia punicea. Recent study from our laboratory has demonstrated that the 3-ODS protects against oxidative toxicity and apoptosis in PC12 cells (Zhang, S.P., Du, X.G., Pu, X.P., 2010. Biol. Pharm. Bull. 33, 1529-1533). The aim of our study is to further investigate the neuroprotective mechanisms of 3-ODS in 1-methyl-4-phenylpyridinium (MPP(+))-induced neurotoxicity in PC12 cells. The results indicated that pre-treatment with 3-ODS significantly increased the cell viability compared with MPP(+) treatment. It also alleviated the oxidative stress by increasing superoxide dismutase (SOD) activity and decreasing malondialdehyde (MDA) level and reactive oxygen specise (ROS) production. Moreover, 3-ODS also attenuated MPP(+)-induced apoptosis by inhibiting Bax and Bcl-2 expressions, activating caspase-9, caspase-3, poly (ADP-ribose) polymerase-1 (PARP-1) cleavage, apoptosis-inducing factor (AIF) translocation and α-synuclein expression. These results suggest that 3-ODS might has applications as a complementary medicine for the treatment of Parkinson's disease (PD) or other neurodegenerative diseases.


Assuntos
1-Metil-4-fenilpiridínio/antagonistas & inibidores , 1-Metil-4-fenilpiridínio/toxicidade , Apoptose/efeitos dos fármacos , Dopaminérgicos/toxicidade , Glucosídeos/farmacologia , Fármacos Neuroprotetores , Estresse Oxidativo/efeitos dos fármacos , Xantonas/farmacologia , Animais , Fator de Indução de Apoptose/metabolismo , Western Blotting , Malondialdeído/metabolismo , Mitocôndrias/efeitos dos fármacos , Células PC12 , Transporte Proteico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Espécies Reativas de Oxigênio , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo , alfa-Sinucleína/antagonistas & inibidores , alfa-Sinucleína/biossíntese , Proteína X Associada a bcl-2/metabolismo
5.
Int J Neuropsychopharmacol ; 16(6): 1341-50, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23195702

RESUMO

Methamphetamine (Meth) abuse can result in long-lasting psychosis and dependence. The nucleus accumbens (NAc), which controls psychomotor and reward behaviours, is an important interface between the limbic system and receives convergent projections from dopaminergic and glutamatergic terminals. This study investigated the involvements of dopaminergic and glutamatergic transmission in the development of Meth psychosis and dependence by using tyrosine hydroxylase heterozygous mutant (TH+/-) mice and N-methyl-d-aspartate receptor knockout (NR2A-/-) mice. Repeated treatment with Meth (1 mg/kg s.c.) for 7 d in wild-type mice led to the development of behavioural abnormalities such as hyperactivity, sensory motor gating deficits and place preference. Associated with the behavioural changes, repeated treatment with Meth led to protein kinase A activation and phosphorylation of Ca2+/calmodulin kinase II and cyclic AMP response element binding protein in the NAc. In contrast, TH+/- and NR2A-/- mice displayed neither behavioural abnormalities nor activation of intracellular signalling pathways in the NAc. These results suggest that both dopaminergic and glutamatergic transmission play a crucial role in the development of Meth psychosis and dependence, which are associated with convergent activation of intracellular signalling pathways in the NAc.


Assuntos
Dopaminérgicos/toxicidade , Dopamina/metabolismo , Ácido Glutâmico/metabolismo , Metanfetamina/toxicidade , Núcleo Accumbens/metabolismo , Transtornos Psicóticos/etiologia , Transtornos Psicóticos/patologia , Estimulação Acústica/efeitos adversos , Análise de Variância , Animais , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Inibição Psicológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Mutação/genética , Núcleo Accumbens/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/genética , Reflexo de Sobressalto/efeitos dos fármacos , Reflexo de Sobressalto/genética , Tirosina 3-Mono-Oxigenase/deficiência
6.
Fundam Clin Pharmacol ; 25(5): 608-18, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21077938

RESUMO

A number of experimental models of L-DOPA-induced dyskinesia have been proposed, but these models result in a low to medium rate of dyskinetic animals with mild to severe symptoms. The objective of this study was to combine a model of 6-OHDA-induced parkinsonism and of L-DOPA-induced dyskinesia in rats to establish a reliable preclinical model. Two stereotaxic injections of 6-OHDA were administered in the left striatum. This model led to 90-100% of rats with a marked contralateral circling behaviour, significant limb use asymmetry (20%), a decrease in ipsilateral striatal dopamine content (70%) and degeneration of dopamine neurons in the substantia nigra (70%). Chronic treatment with L-DOPA was administered for 35 days and consisted of three phases with incremental daily doses. The third phase resulted in 83-90% of rats developing severe abnormal involuntary movements (AIMs) which included limb and locomotive dyskinesia, axial dystonia and orolingual dyskinesia. Reproducibility of the model, criteria of strict blinding, placebo-controlled design, randomization of study subjects and pretrial determination of sample size were used to measure efficacy of amantadine and istradefylline and to validate the protocol design. Acute or subchronic post-treatment with amantadine reduced the severity of dyskinesia while istradefylline punctually attenuated AIMs. Our experimental conditions using gradual development of dyskinesia induced by increasing doses of L-DOPA resulted in a reliable model of L-DOPA-induced dyskinesia with a high rate of dyskinetic rats.


Assuntos
Amantadina/uso terapêutico , Antiparkinsonianos/uso terapêutico , Modelos Animais de Doenças , Discinesia Induzida por Medicamentos/tratamento farmacológico , Transtornos Parkinsonianos/tratamento farmacológico , Purinas/uso terapêutico , Antagonistas do Receptor A2 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/toxicidade , Amantadina/farmacologia , Amantadina/toxicidade , Animais , Antiparkinsonianos/efeitos adversos , Antiparkinsonianos/farmacologia , Comportamento Animal , Benserazida/farmacologia , Bioensaio , Corpo Estriado , Dopamina/fisiologia , Dopaminérgicos/farmacologia , Dopaminérgicos/toxicidade , Esquema de Medicação , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Levodopa/efeitos adversos , Levodopa/farmacologia , Levodopa/toxicidade , Masculino , Oxidopamina/toxicidade , Transtornos Parkinsonianos/induzido quimicamente , Placebos , Purinas/farmacologia , Purinas/toxicidade , Distribuição Aleatória , Ratos , Reprodutibilidade dos Testes , Rotação , Tamanho da Amostra , Método Simples-Cego , Simpatolíticos/toxicidade
7.
Behav Brain Res ; 207(2): 387-93, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-19857526

RESUMO

Cognitive deficits are a core feature of patients with methamphetamine (METH) abuse. It has been reported that repeated METH treatment impairs long-term recognition memory in the novel object recognition test (NORT) in mice. Recent studies indicate that silibinin, a flavonoid derived from the herb milk thistle, has potent neuroprotective effects in cell cultures and several animal models of neurological diseases. However, its effect on the cognitive deficit induced by METH remains unclear. In the present study, we attempt to clarify the effect of silibinin on impairments of recognition memory caused by METH in mice. Mice were co-administered silibinin with METH for 7 days and then cognitive function was assessed by NORT after 7-day withdrawal. Tissue levels of dopamine and serotonin as well as their metabolites in the prefrontal cortex and hippocampus were measured 1 day after NORT. Silibinin dose-dependently ameliorated the impairment of recognition memory caused by METH treatment in mice. Silibinin significantly attenuated the decreases in the dopamine content of the prefrontal cortex and serotonin content of the hippocampus caused by METH treatment. We also found a correlation between the recognition values and dopamine and serotonin contents of the prefrontal cortex and hippocampus. The effect of silibinin on cognitive impairment may be associated with an amelioration of decreases in dopamine and serotonin levels in the prefrontal cortex and hippocampus, respectively. These results suggest that silibinin may be useful as a pharmacological tool to investigate the mechanisms of METH-induced cognitive impairments.


Assuntos
Encéfalo/efeitos dos fármacos , Dopaminérgicos/toxicidade , Transtornos da Memória/tratamento farmacológico , Metanfetamina/toxicidade , Nootrópicos/farmacologia , Animais , Encéfalo/metabolismo , Cognição/efeitos dos fármacos , Cognição/fisiologia , Dopamina/metabolismo , Dopaminérgicos/administração & dosagem , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/metabolismo , Metanfetamina/administração & dosagem , Camundongos , Camundongos Endogâmicos ICR , Nootrópicos/administração & dosagem , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Serotonina/metabolismo , Silibina , Silimarina/administração & dosagem , Silimarina/farmacologia
8.
Curr Eye Res ; 34(9): 785-90, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19839872

RESUMO

PURPOSE: To investigate whether systemic administration of methamphetamine (METH) induces retinal damage in CD1 mice. MATERIALS AND METHODS: Eighteen male CD1 mice were randomly assigned to three groups, six mice per group: Group 1 receiving a single dose of 40 mg/kg METH, Group 2 receiving four doses of 10 mg/kg METH, and Group 3 (control) receiving 40 mg/kg 0.9% NaCl solution. METH and NaCl were administered by intraperitoneal injection. Immunostaining of glial fibrillary acidic protein (GFAP), S-100 for astrocytes and Muller cells, CD11b for microglia, and tyrosine hydroxylase (TH) and TUNEL labeling for apoptotic cell death were performed on the retinal sections on day 1 and day 7 post-exposure. RESULTS: GFAP and S-100 immunoreactivity was observed in Group 1 mice. CD11b+ cells in Group 1 mice showed more intensely stained shorter and thicker cellular processes than Groups 2 and 3, indicating activated microglia in the mice exposed to large-dose METH. No significant difference in TH level was seen among the three groups. TUNEL labeling did not reveal positive cells in the retinas of any of the 18 CD1 mice. CONCLUSIONS: A single large dose of METH induces an increase in short-term protein expression of GFAP and S-100 and in microglial activation. The results suggest that METH has a neurotoxic effect on CD1 mouse retina.


Assuntos
Dopaminérgicos/toxicidade , Metanfetamina/toxicidade , Retina/efeitos dos fármacos , Doenças Retinianas/induzido quimicamente , Animais , Apoptose , Antígeno CD11b/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Técnicas Imunoenzimáticas , Marcação In Situ das Extremidades Cortadas , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Retina/metabolismo , Retina/patologia , Doenças Retinianas/metabolismo , Doenças Retinianas/patologia , Proteínas S100/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
9.
Neurochem Int ; 54(7): 447-51, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19428787

RESUMO

Striatal administration of 1-methyl-4-phenylpyridinium (MPP(+)), the active metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), causes nigrostriatal dopaminergic pathway damage similar to that observed in Parkinson's disease. Copper acts as a prosthetic group of several antioxidant enzymes and recent data show that copper attenuated MPP(+)-evoked neurotoxicity. We evaluated the effect of copper (as a supplement) upon proteins nitration (60 kDa) and tyrosine hydroxylase (TH) inactivation induced by MPP(+) (10 microg/8 microL) injection into the rat striatum. Copper pretreatment (10 micromol/kg i.p.) prevented both MPP(+)-induced proteins nitration and TH inactivation. Copper treatment also prevented the dopamine-depleting effect of MPP(+) injection. Those results were accompanied by a significant reduction of enzymatic activity of the constitutive nitric oxide synthase (cNOS), whereas, the protein levels of the three isoforms of NOS remained unchanged. Results indicate that the effect of copper against MPP(+)-induced proteins nitration and TH inactivation in the striatum of rat may be mediated by a reduction of cNOS activity.


Assuntos
Sulfato de Cobre/farmacologia , Intoxicação por MPTP/prevenção & controle , Neostriado/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nitratos/metabolismo , Tirosina 3-Mono-Oxigenase/antagonistas & inibidores , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Western Blotting , Dopamina/metabolismo , Dopaminérgicos/toxicidade , Eletroforese em Gel de Poliacrilamida , Masculino , Neostriado/efeitos dos fármacos , Óxido Nítrico Sintase Tipo I/biossíntese , Doença de Parkinson Secundária/prevenção & controle , Ratos , Ratos Wistar
10.
Pharmacol Biochem Behav ; 90(3): 363-71, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18485464

RESUMO

Common flowering quince (FQ) is the fruit of Chaenomeles speciosa (Sweet) Nakai. FQ-containing cocktails have been applied to the treatment of neuralgia, migraine, and depression in traditional Chinese medicine. The present study assessed whether FQ is effective in dopamine transporter (DAT) regulation and antiparkinsonism by utilizing in vitro and in vivo assays, respectively. FQ at concentrations of 1-1000 microg/ml concentration-dependently inhibited dopamine uptake by Chinese hamster ovary (CHO) cells stably expressing DAT (D8 cells) and by synaptosomes. FQ had a slight inhibitory action on norepinephrine uptake by CHO cells expressing the norepinephrine transporter and no inhibitory effect on gamma-aminobutyric acid (GABA) uptake by CHO cells expressing GABA transporter-1 or serotonin uptake by the serotonin transporter. A viability assay showed that FQ mitigated 1-methyl-4-phenylpyridinium-induced toxicity in D8 cells. Furthermore, in behavioral studies, FQ alleviated rotational behavior in 6-hydroxydopamine-treated rats and improved deficits in endurance performance in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. Furthermore, immunohistochemistry revealed that FQ markedly reduced the loss of tyrosine hydroxylase-positive neurons in the substantia nigra in MPTP-treated mice. In summary, FQ is a selective, potent DAT inhibitor and has antiparkinsonian-like effects that are mediated possibly by DAT suppression. FQ has the potential to be further developed for Parkinson's disease treatment.


Assuntos
Antiparkinsonianos , Proteínas da Membrana Plasmática de Transporte de Dopamina/antagonistas & inibidores , Rosaceae/química , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Dopamina/metabolismo , Dopaminérgicos/toxicidade , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Inibidores da Captação de Dopamina/farmacologia , Tratos Extrapiramidais/efeitos dos fármacos , Frutas/química , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/genética , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Organismos Geneticamente Modificados , Extratos Vegetais/farmacologia , Equilíbrio Postural/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
11.
Pediatr Res ; 62(1): 14-9, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17515828

RESUMO

The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) inhibits the mitochondrial complex I of the respiratory chain. This results in ATP and ion homeostasis disturbances, which lead to selective death of the substantia nigra dopaminergic neurons. Well known as a Parkinson's disease model, the MPTP animal model also provides a potential paradigm of the energy deficiencies found in childhood. In these conditions, anticonvulsants may provide neuroprotection by limiting cellular energy consumption. We tested valproate, topiramate and lamotrigine in the MPTP mouse model. Dopamine transporter (DAT) density was assessed by quantitative autoradiography, tyrosine hydroxylase (TH) was evaluated by immunohistochemistry and dopamine (DA) levels by HPLC-ED whereas neuronal apoptosis was monitored through active caspase-3. Expectedly, the DAT density, TH immunoreactive neurons and DA content in the MPTP group were respectively reduced to 51%, 40% and 26% versus control animals. Unlike valproate and topiramate, lamotrigine provided a significant neuroprotection against MPTP in maintaining these levels at 99%, 74% and 58% respectively and reducing the induced apoptosis. Altogether, the data indicate that lamotrigine limits dopaminergic neuronal death in the substantia nigra and promotes striatal dendrites sprouting. Lamotrigine, a widely used and well-tolerated molecule in young patients, could represent a valuable adjuvant therapy in various energy deficiency conditions during childhood.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/metabolismo , Anticonvulsivantes/metabolismo , Dopaminérgicos/metabolismo , Metabolismo Energético , Fármacos Neuroprotetores/metabolismo , Neurotoxinas/metabolismo , Triazinas/metabolismo , Animais , Anticonvulsivantes/química , Apoptose/fisiologia , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Dopamina/metabolismo , Dopaminérgicos/toxicidade , Frutose/análogos & derivados , Frutose/metabolismo , Lamotrigina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/química , Neurotoxinas/toxicidade , Topiramato , Triazinas/química , Ácido Valproico/metabolismo
12.
Neurochem Res ; 32(6): 1071-80, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17401669

RESUMO

The neurotoxin 1-methy-4-phenylpyridinium (MPP(+)) is used for its' capacity to induce Parkinsonism through its inhibitory effects on mitochondrial complex I. This inhibition disrupts cellular energy formation and aerobic glycolysis. The objective of this study was to demonstrate that the toxic effect of mitochondrial aerobic pathway inhibition with MPP(+ )can be reduced by stimulating anaerobic glycolysis using glucose supplementation. In this study, C6 Glioma cell viability was examined in the presence of different concentrations of MPP alone and with the addition of glucose. The results obtained indicate that there was a significant increase (P < 0.001) in cell viability in cells treated with glucose and MPP(+ )verses cells treated with MPP(+ )alone. Fluorometric analysis using 100 microM Rhodamine 123 indicated mitochondrial membrane potential was not restored in MPP(+ )treated cells with glucose; however, normal cell viability was confirmed using 2 microg/ml Fluorescein diacetate. This dual fluorescence indicated mitochondrial damage from MPP(+ )while glucose augmented cell survival. Further confirmation of cell survival upon damage to the mitochondria was evident in TUNEL staining. Positive staining was prominent only in MPP(+) treatment groups alone, while control and co-treated groups exhibited little to no TUNEL staining. ATP measurements of all MPP(+) treated groups exhibited a significant (P < 0.001) decrease verses control. Groups co-treated with MPP(+ )and glucose revealed a significant increase (250 microM group: P < 0.001) in ATP. It was concluded from this study that glucose supplementation was able to sustain cellular viability and ATP production through anaerobic glycolysis despite the inhibitory effect of MPP(+ )on aerobic glycolysis.


Assuntos
1-Metil-4-fenilpiridínio/antagonistas & inibidores , 1-Metil-4-fenilpiridínio/toxicidade , Neoplasias Encefálicas/metabolismo , Dopaminérgicos/toxicidade , Glioma/metabolismo , Glicólise/efeitos dos fármacos , Trifosfato de Adenosina/biossíntese , Anaerobiose , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Fluoresceína , Corantes Fluorescentes , Glucose/metabolismo , Marcação In Situ das Extremidades Cortadas , Luminescência , Potenciais da Membrana/efeitos dos fármacos , Microscopia de Fluorescência , Mitocôndrias/efeitos dos fármacos , Ratos
13.
Neuropsychopharmacology ; 32(3): 531-41, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16760923

RESUMO

Some of the major concerns related to methamphetamine (METH) abuse are the neuronal damage inflicted at dopamine (DA) nerve terminals and the cognitive deficits observed in human METH abusers. We have shown that a high dose of METH selectively depleted dopaminergic markers in striatum, frontal cortex and amygdala of Swiss Webster mice, and impaired learned place preference. In this study, we investigated whether deficits in consolidation of place learning, as a consequence of METH neurotoxicity, underlie the underperformance of cocaine conditioned place preference (CPP). Administration of METH (5 mg/kg x 3) to Swiss Webster mice decreased striatal tyrosine hydroxylase (TH) immunoreactive neurons and significantly increased glial fibrillary acidic protein (GFAP) expression, confirming the neurotoxic potential of METH in mice. This treatment significantly attenuated the establishment of cocaine (15 mg/kg) CPP compared to control. To investigate whether manipulation of the consolidation phase improves learned place preference, mice were trained by cocaine and received daily post-training injections of DA receptor agonists or N-acetylcysteine (NAC). As memory consolidation occurs shortly after training, drugs were administered either immediately or 2 h post-training. Immediate post-training administration of the D1 DA receptor agonist SKF38393 (5, 10, and 20 mg/kg) or the D2 DA receptor agonist quinpirole (0.25, 0.5, and 1.0 mg/kg) did not improve the establishment of CPP following METH neurotoxicity. However, immediate but not delayed NAC administration (50 and 100 mg/kg) enhanced cocaine CPP following METH neurotoxicity and had no effect on control CPP. The levels of the reduced form of glutathione (GSH) in striatum, amygdala, hippocampus and frontal cortex were significantly lower in METH-treated mice compared to control during the period of CPP training. Acute and repeated administration of NAC to METH-treated mice restored the decreased brain GSH but had no effect on controls. Results suggest that METH-induced dopaminergic neurotoxicity is associated with impairment of consolidation of learned place preference, and that this impairment is improved by immediate post-training administration of the glutathione precursor NAC and not by D1 or D2 DA receptor agonists. Restoration of brain glutathione levels immediately post-training may facilitate the consolidation process.


Assuntos
Acetilcisteína/uso terapêutico , Condicionamento Operante/efeitos dos fármacos , Dopamina/metabolismo , Sequestradores de Radicais Livres/uso terapêutico , Deficiências da Aprendizagem/tratamento farmacológico , Síndromes Neurotóxicas/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Química Encefálica/efeitos dos fármacos , Cocaína/administração & dosagem , Modelos Animais de Doenças , Dopaminérgicos/toxicidade , Agonistas de Dopamina/uso terapêutico , Inibidores da Captação de Dopamina/administração & dosagem , Relação Dose-Resposta a Droga , Interações Medicamentosas , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Glutationa/metabolismo , Deficiências da Aprendizagem/etiologia , Masculino , Metanfetamina/toxicidade , Camundongos , Síndromes Neurotóxicas/complicações , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/etiologia , Tirosina 3-Mono-Oxigenase/metabolismo
14.
Ann N Y Acad Sci ; 1074: 261-71, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17105922

RESUMO

Repeated methamphetamine (METH) administration to animals can result in long-lasting decreases in brain dopamine (DA) and serotonin (5-HT) content. Calcitriol, the active metabolite of vitamin D, has potent effects on brain cells, both in vitro and in vivo, including the ability to upregulate trophic factors and protect against various lesions. The present experiments were designed to examine the ability of calcitriol to protect against METH-induced reductions in striatal and nucleus accumbens levels of DA and 5-HT. Male Fischer-344 rats were administered vehicle or calcitriol (1 microg/kg, s.c.) once a day for eight consecutive days. After the seventh day of treatment the animals were given METH (5 mg/kg, s.c.) or saline four times in 1 day at 2-h intervals. Seven days later the striata and accumbens were harvested from the animals for high-performance liquid chromatography (HPLC) analysis of monoamines and metabolites. In animals treated with vehicle and METH, there were significant reductions in DA, 5-HT, and their metabolites in both the striatum and accumbens. In animals treated with calcitriol and METH, the magnitude of the METH-induced reductions in DA, 5-HT, and metabolites was substantially and significantly attenuated. The calcitriol treatments did not reduce the hyperthermia associated with multiple injections of METH, indicating that the neuroprotective effects of calcitriol are not due to the prevention of increases in body temperature. These results suggest that calcitriol can provide significant protection against the DA- and 5-HT-depleting effects of neurotoxic doses of METH.


Assuntos
Calcitriol/farmacologia , Dopaminérgicos/toxicidade , Dopamina/metabolismo , Metanfetamina/toxicidade , Serotonina/metabolismo , Animais , Corpo Estriado/metabolismo , Interações Medicamentosas , Hipertermia Induzida , Masculino , Núcleo Accumbens/metabolismo , Ratos , Ratos Endogâmicos F344
15.
Behav Pharmacol ; 17(5-6): 393-402, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16940760

RESUMO

In spite of the current availability of several pharmacological therapies for the treatment of Parkinson's disease, side effects are invariably manifested during long-term treatment. Dyskinesia, wearing-off and on-off are among the most disabling side effects produced by the dopamine precursor L-dihydroxyphenylalanine and, to a lesser degree, by other pharmacological treatments based on dopamine receptor agonism. Evaluation of the side effects, in particular dyskinesia, produced by antiparkinsonian drug treatments, therefore represents a critical issue in drug validation prior to a clinical trial. Moreover, a reliable model of dyskinesia is a fundamental requirement for the study of the as yet unknown mechanisms at the basis of this severely disabling side effect. The present review aims to provide a critical evaluation of the validity, reliability and utility of animal models of dyskinesia. In the first part of this review, we present a brief overview of the different models of Parkinson's disease focusing on those utilized for the evaluation of dyskinetic movements, then proceed to critically examine the turning behaviour model in an attempt to assess the way in which it has influenced the evaluation of drugs utilized in the treatment of Parkinson's disease. Subsequently, the various models of dyskinesia are reviewed and conclusions are drawn as to how the environment in which experiments are performed can influence the behaviour observed.


Assuntos
Antiparkinsonianos/toxicidade , Comportamento Animal/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Discinesia Induzida por Medicamentos/etiologia , Transtornos Parkinsonianos/tratamento farmacológico , Animais , Dopaminérgicos/toxicidade , Levodopa/toxicidade , Atividade Motora/efeitos dos fármacos , Destreza Motora/efeitos dos fármacos , Comportamento Estereotipado/efeitos dos fármacos
16.
Neuropathol Appl Neurobiol ; 32(4): 428-40, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16866988

RESUMO

Iron is potentially toxic to oligodendrocyte progenitors due to its high intracellular levels and its ability to catalyse oxidant-producing reactions. Oxidative stress resulting from a hypoxic-ischaemic insult has been implicated in death of oligodendrocyte progenitors that occurs in the hypomyelinating disorder periventricular leucomalacia. Ischaemic insults induce the release of various neurotransmitters, including dopamine (DA), and we previously showed that DA is toxic to cultured oligodendrocytes, by inducing oxidative stress and apoptosis. Therefore, we investigated the role of iron in DA-induced cell death in oligodendrocyte progenitors. Intracellular iron levels were altered using an iron chelator, deferoxamine (DFO), and supplementation with ferrous sulphate (FeSO(4)). Addition of FeSO(4) to cultures increased DA-induced toxicity as assessed by mitochondrial dehydrogenase activity and cellular release of lactate dehydrogenase. Furthermore, FeSO(4) increased expression of the stress protein heme oxygenase-1 (HO-1), nuclear condensation and caspase-3 activation. In contrast, preincubation with DFO reduced these events as well as cleavage of alpha-spectrin, a caspase-3 substrate. In addition, FeSO(4) reversed the protective effect of DFO on DA-induced cytotoxicity, HO-1 expression and caspase-3 activation. These results indicate that elevated levels of free iron contribute to DA-induced toxicity in oligodendrocyte progenitors.


Assuntos
Dopaminérgicos/toxicidade , Dopamina/toxicidade , Compostos Ferrosos/toxicidade , Oligodendroglia/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3 , Caspases/efeitos dos fármacos , Caspases/metabolismo , Núcleo Celular/efeitos dos fármacos , Desferroxamina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Compostos Ferrosos/metabolismo , Heme Oxigenase-1/efeitos dos fármacos , Necrose , Oligodendroglia/patologia , Ratos , Ratos Sprague-Dawley , Sideróforos/farmacologia , Células-Tronco/patologia
17.
Neurodegener Dis ; 1(4-5): 175-83, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-16908987

RESUMO

The neurotoxin MPTP and its active metabolite MPP+ cause Parkinson's disease (PD)-like symptoms in vertebrates by selectively destroying dopaminergic neurons in the substantia nigra. MPTP/MPP+ models have been established in rodents to screen for pharmacologically active compounds. In addition to being costly and time consuming, these animal models are not suitable for large scale testings using compound libraries. We present a novel MPP+-based model for high-throughput screenings using the nematode Caenorhabditis elegans. Incubation of C. elegans with MPTP or its active metabolite MPP+ resulted in strong symptomatic defects including reduced mobility and increased lethality, and is correlated with a specific degeneration of the dopaminergic neurons. The phenotypic consequences of MPTP/MPP+ treatments were recorded using automated hardware and software for quantification. Incubation of C. elegans with a variety of pharmacologically active components used in PD treatment reduced the MPP+-induced defects. Our data suggest that the C. elegans MPTP/MPP+ model can be used for the quantitative evaluation of anti-PD drugs.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Antiparkinsonianos/farmacologia , Caenorhabditis elegans/fisiologia , Dopaminérgicos/toxicidade , Doença de Parkinson Secundária/induzido quimicamente , Animais , Caenorhabditis elegans/enzimologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Proteínas de Fluorescência Verde/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fenótipo , Rotenona/toxicidade , Desacopladores/toxicidade
18.
Exp Neurol ; 184(1): 521-9, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14637121

RESUMO

The herbal remedy, ginseng, has recently been demonstrated to possess neurotrophic and neuroprotective properties, which may be useful in preventing various forms of neuronal cell loss including the nigrostriatal degeneration seen in Parkinson's disease (PD). In these studies, we examine the potential neuroprotective actions of the ginseng extract, G115, in two rodent models of PD. Animals received oral administration of G115 prior to and/or following exposure to the parkinsonism-inducing neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), in mice, or its toxic metabolite, 1-methyl-4-phenylpyridinium (MPP(+)), in rats. Such treatment significantly and dramatically blocked tyrosine hydroxylase-positive cell loss in the substantia nigra and reduced the appearance of locomotor dysfunction. Thus, oral administration of ginseng appears to provide protection against neurotoxicity in rodent models of PD. Further examination of the neuroprotective actions of ginseng and its various elements may provide a potential means of slowing the progress of PD.


Assuntos
Fármacos Neuroprotetores , Panax/química , Doença de Parkinson Secundária/prevenção & controle , Fitoterapia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/antagonistas & inibidores , Animais , Dopaminérgicos/toxicidade , Relação Dose-Resposta a Droga , Feminino , Imuno-Histoquímica , Locomoção/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Doença de Parkinson Secundária/patologia , Doença de Parkinson Secundária/fisiopatologia , Extratos Vegetais/uso terapêutico , Compostos de Piridínio/toxicidade , Ratos , Ratos Sprague-Dawley
19.
Neurotoxicology ; 23(3): 367-74, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12387363

RESUMO

In animal models of Parkinson's disease (PD), the toxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is mediated by oxidative stress, especially by nitric oxide (NO). Inhibition of NO synthase (NOS) activity in the brain produces a neuroprotective effect against PD induced by MPTP Green tea containing high levels of (-)-epigallocatechin 3-gallate (EGCG) was administered to test whether EGCG attenuates MPTP-induced PD in mice through the inhibition of NOS expression. Both tea and the oral administration of EGCG prevented the loss of tyrosine hydroxylase (TH)-positive cells in the substantia nigra (SN) and of TH activity in the striatum. These treatments also preserved striatal levels of dopamine and its metabolites, 3,4-dihydroxyphenylacetic acid and homovanillic acid (HVA). Both tea and EGCG decreased expressions of nNOS in the substantia nigra. Also tea plus MPTP and EGCG plus MPTP treatments decreased expressions of neuronal NO synthase (nNOS) at the similar levels of EGCG treatment group. Therefore, the preventive effects of tea and EGCG may be explained by the inhibition of nNOS in the substantia nigra.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/antagonistas & inibidores , Catequina/análogos & derivados , Catequina/farmacologia , Dopaminérgicos/toxicidade , Óxido Nítrico/fisiologia , Doença de Parkinson Secundária/prevenção & controle , Chá/química , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Western Blotting , Ácido Homovanílico/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo I , Doença de Parkinson Secundária/induzido quimicamente , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Substância Negra/efeitos dos fármacos , Substância Negra/enzimologia , Tirosina 3-Mono-Oxigenase/metabolismo
20.
Brain Res ; 862(1-2): 247-52, 2000 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-10799693

RESUMO

The present study was designed to understand the role of an antioxidant, selenium (Se) on methamphetamine (MA)-induced dopaminergic cell damage in the substantia nigra (SN). Male C57BL/6J mice were fed either selenium-deficient (<0.01 ppm Se) or selenium-replete (0.2 ppm Se) diet for 90 days. Se-deficiency potentiates MA-induced reductions of tyrosine hydroxylase-like immunoreactivity (TH-IR), dopamine (DA) and its metabolites, 3, 4-dihydroxyphenylacetic acid (DOPAC) and homovanilic acid (HVA) in the SN. These dopaminergic toxicities were comparable to that induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). By contrast, Se-repletion significantly blocked dopaminergic toxicity after MA treatments. These results suggest that Se-deficient MA-treated mouse is a relevant model of Parkinsonism, and that optimal level of Se plays a crucial role in preventing nigral dopaminergic toxicity induced by MA. However, different mechanisms in the thermoregulation mediated by MA or MPTP remain to be further determined.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Dopaminérgicos/toxicidade , Metanfetamina/toxicidade , Degeneração Neural/induzido quimicamente , Selênio/deficiência , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Antioxidantes/metabolismo , Dieta , Dopamina/metabolismo , Ácido Homovanílico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Substância Negra/citologia , Substância Negra/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA