Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Neurobiol ; 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38289456

RESUMO

Epilepsy is characterized by a sustained depolarization and repeated discharge of neurons, attributed to overstimulation of N-methyl-D-aspartate receptors (NMDAr). Herein, we propose that probenecid (PROB), an inhibitor of the activity of some ATP binding-cassette transporters (ABC-transporters) can modify NMDAr activity and expression in amygdaloid kindled model. Some studies have suggested that NMDAr expression could be regulated by inhibiting the activity of P-glycoprotein (MDR1) and drug resistance protein-1 (MRP1). Besides, PROB was found to interact with other proteins with proven activity in the kindling model, such as TRPV2 channels, OAT1, and Panx1. Administering PROB at two doses (100 and 300 mg/kg/d) for 5 d decreased after-discharge duration and Racine behavioral scores. It also reduced the expression of NR2B and the activity of total NOS and the expression of nNOS with respect to the kindling group. In a second protocol, voltage-clamp measurements of NMDA-evoked currents were performed in CA1 hippocampal cells dissociated from control and kindled rats. PROB produced a dose-dependent reduction in NMDA-evoked currents. In neurons from kindled rats, a residual NMDA-evoked current was registered with respect to control animals, while a reduction in NMDA-evoked currents was observed in the presence of 20 mM PROB. Finally, we evaluated the expression of MRP1 and MDR1 in order to establish a relationship between the reduction of kindling parameters, the inhibition of NMDA-type currents, and the expression of these transporters. Based on our results, we conclude that at the concentrations used, PROB inhibits currents evoked by NMDA in dissociated neurons of control and kindled rats. In the kindling model, at the tested doses, PROB decreases the after-discharge duration and Racine behavioral score in the kindling model. We propose a mechanism that could be dependent on the expression of ABC-type transporters.

2.
Rev Neurosci ; 33(7): 819-827, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-35411760

RESUMO

Huntington's disease (HD), a neurodegenerative disorder caused by an expansion of the huntingtin triplet (Htt), is clinically characterized by cognitive and neuropsychiatric alterations. Although these alterations appear to be related to mutant Htt (mHtt)-induced neurotoxicity, several other factors are involved. The gut microbiota is a known modulator of brain-gut communication and when altered (dysbiosis), several complaints can be developed including gastrointestinal dysfunction which may have a negative impact on cognition, behavior, and other mental functions in HD through several mechanisms, including increased levels of lipopolysaccharide, proinflammatory cytokines and immune cell response, as well as alterations in Ca2+ signaling, resulting in both increased intestinal and blood-brain barrier (BBB) permeability. Recently, the presence of dysbiosis has been described in both transgenic mouse models and HD patients. A bidirectional influence between host brain tissues and the gut microbiota has been observed. On the one hand, the host diet influences the composition and function of microbiota; and on the other hand, microbiota products can affect BBB permeability, synaptogenesis, and the regulation of neurotransmitters and neurotrophic factors, which has a direct effect on host metabolism and brain function. This review summarizes the available evidence on the pathogenic synergism of dysbiosis and homocysteine, and their role in the transgression of BBB integrity and their potential neurotoxicity of HD.


Assuntos
Doença de Huntington , Animais , Citocinas , Disbiose , Homocisteína , Humanos , Lipopolissacarídeos , Camundongos , Fatores de Crescimento Neural
3.
Front Immunol ; 13: 1039427, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36591299

RESUMO

In the past two years, the world has faced the pandemic caused by the severe acute respiratory syndrome 2 coronavirus (SARS-CoV-2), which by August of 2022 has infected around 619 million people and caused the death of 6.55 million individuals globally. Although SARS-CoV-2 mainly affects the respiratory tract level, there are several reports, indicating that other organs such as the heart, kidney, pancreas, and brain can also be damaged. A characteristic observed in blood serum samples of patients suffering COVID-19 disease in moderate and severe stages, is a significant increase in proinflammatory cytokines such as interferon-α (IFN-α), interleukin-1ß (IL-1ß), interleukin-2 (IL-2), interleukin-6 (IL-6) and interleukin-18 (IL-18), as well as the presence of autoantibodies against interferon-α (IFN-α), interferon-λ (IFN-λ), C-C motif chemokine ligand 26 (CCL26), CXC motif chemokine ligand 12 (CXCL12), family with sequence similarity 19 (chemokine (C-C motif)-like) member A4 (FAM19A4), and C-C motif chemokine ligand 1 (CCL1). Interestingly, it has been described that the chronic cytokinemia is related to alterations of blood-brain barrier (BBB) permeability and induction of neurotoxicity. Furthermore, the generation of autoantibodies affects processes such as neurogenesis, neuronal repair, chemotaxis and the optimal microglia function. These observations support the notion that COVID-19 patients who survived the disease present neurological sequelae and neuropsychiatric disorders. The goal of this review is to explore the relationship between inflammatory and humoral immune markers and the major neurological damage manifested in post-COVID-19 patients.


Assuntos
Doenças Neurodegenerativas , Síndrome de COVID-19 Pós-Aguda , Humanos , Quimiocinas , COVID-19 , Imunidade , Interferon-alfa , Interleucina-6 , Ligantes , Síndrome de COVID-19 Pós-Aguda/complicações , Síndrome de COVID-19 Pós-Aguda/imunologia , Síndrome de COVID-19 Pós-Aguda/fisiopatologia , SARS-CoV-2 , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/fisiopatologia
4.
Neurosci Biobehav Rev ; 126: 361-367, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33819547

RESUMO

Lead (Pb) is a pollutant commonly found in the environment, despite the implementation of public health policies intended to remove it. Due to its chemical characteristics as a divalent ion, Pb interacts with cells, enzymes, and tissues, causing pathological, physical, and behavioral alterations. Recent biotechnological advances have helped us to understand the mechanisms underlying the damage caused by Pb in human populations and in experimental models, and new evidence on the epigenetic alterations caused by exposition to environmental Pb is available. It is known that Pb exposure impacts on behavior (causing aggressiveness, anxiety, and depression), leading to learning deficit and locomotor activity alterations, and its presence has been linked with the abnormal release of neurotransmitters and other biochemical changes involved in these disorders. Still, further reductionist studies are required to determine the effects of Pb exposure on DNA and protein expression and understand the processes underlying the diseases caused by Pb. This will also indicate possible therapeutic targets to offset the negative effects of the heavy metal. By elucidating the epigenetic changes involved, it would be possible to manipulate them and propose novel therapeutic approaches in this area. This review is aimed to provide an overview of studies that link Pb exposure to behavioral changes, as well as biochemical and epigenetic alterations at a neurotransmitter level, considering the importance of this metal in behavior abnormalities.


Assuntos
Epigênese Genética , Chumbo , Ansiedade , Expressão Gênica , Humanos , Chumbo/toxicidade , Processamento de Proteína Pós-Traducional
5.
Nutr Neurosci ; 24(1): 1-12, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30822260

RESUMO

Background: Essential fatty acids (EFAs) and non-essential fatty acids (nEFAs) exert experimental and clinical neuroprotection in neurodegenerative diseases. The main EFAs, eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), nEFAs, and oleic acid (OA) contained in olive and fish oils are inserted into the cell membranes, but the exact mechanism through which they exert neuroprotection is still unknown. Objectives and Methods: In this study, we assessed the fatty acids content and membrane fluidity in striatal rat synaptosomes after fatty acid-rich diets (olive- or a fish-oil diet, 15% w/w). Then, we evaluated the effect of enriching striatum synaptosomes with fatty acids on the oxidative damage produced by the prooxidants ferrous sulfate (FeSO4) or quinolinic acid (QUIN). Results and Discussion: Lipid profile analysis in striatal synaptosomes showed that EPA content increased in the fish oil group in comparison with control and olive groups. Furthermore, we found that synaptosomes enriched with fatty acids and incubated with QUIN or FeSO4 showed a significant oxidative damage reduction. Results suggest that EFAs, particularly EPA, improve membrane fluidity and confer antioxidant effect.


Assuntos
Membrana Celular/metabolismo , Corpo Estriado/metabolismo , Ácidos Graxos/metabolismo , Estresse Oxidativo , Sinaptossomos/metabolismo , Animais , Membrana Celular/ultraestrutura , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/ultraestrutura , Ácidos Graxos/administração & dosagem , Óleos de Peixe/administração & dosagem , Masculino , Óleos de Plantas/administração & dosagem , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Sinaptossomos/ultraestrutura
6.
Mov Disord ; 35(7): 1113-1127, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32410324

RESUMO

Huntington's disease is an autosomal-dominant, neurodegenerative disorder caused by a CAG repeat expansion in exon-1 of the huntingtin gene. Alterations in cholesterol metabolism and distribution have been reported in Huntington's disease, including abnormal interactions between mutant huntingtin and sterol regulatory element-binding proteins, decreased levels of apolipoprotein E/cholesterol/low-density lipoprotein receptor complexes, and alterations in the synthesis of ATP-binding cassette transporter A1. Plasma levels of 24S-hydroxycholestrol, a key intermediary in cholesterol metabolism and a possible marker in neurodegenerative diseases, decreased proportionally to the degree of caudate nucleus atrophy. The interaction of mutant huntingtin with sterol regulatory element-binding proteins is of particular interest given that sterol regulatory element-binding proteins play a dual role: They take part in lipid and cholesterol metabolism, but also in the inflammatory response that induces immune cell migration as well as toxic effects, particularly in astrocytes. This work summarizes current evidence on the metabolic and immune implications of sterol regulatory element-binding protein dysregulation in Huntington's disease, highlighting the potential use of drugs that modulate these alterations. © 2020 International Parkinson and Movement Disorder Society.


Assuntos
Encéfalo/metabolismo , Colesterol/metabolismo , Doença de Huntington/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Humanos , Doença de Huntington/genética , Metabolismo dos Lipídeos
7.
Behav Brain Res ; 366: 108-117, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-30898683

RESUMO

Nitric oxide (NO) plays a leading role in learning and memory processes. Previously, we showed its ability to modify the deleterious effect of immunotoxin 192 IgG-saporin (192-IgG-SAP) in the cholinergic system. The aim of this study was to analyze the potential of a NO donor (molsidomine, MOLS) to prevent the recognition memory deficits resulting from the septal cholinergic denervation by 192 IgG-SAP in rats. Quantification of neuronal and endothelial nitric oxide synthase (nNOS and eNOS, respectively) expression was evaluated in striatum, prefrontal cortex, and hippocampus. In addition, a choline acetyltransferase immunohistochemical analysis was performed in medial septum and assessed the effect of MOLS treatment on the spatial working memory of rats through a recognition memory test. Results showed that 192-IgG-SAP reduced the immunoreactivity of cholinergic septal neurons (41%), compared with PBS-receiving control rats (p < 0.05). Treatment with MOLS alone failed to antagonize the septal neuron population loss but prevented the progressive abnormal morphological changes of neurons. Those animals exposed to 192-IgG-SAP immunotoxin exhibited a reduction of cortical nNOS expression against the control group, whereas expression was enhanced in the 192-IgG-SAP + MOLS group. The most relevant finding was the recovering of the discrimination index exhibited by the 192-IgG-SAP + MOLS group. When compared with the rats exposed to the 192-IgG-SAP immunotoxin, they reached values similar to those observed in the PBS group. Our results show that although MOLS failed to block the cholinergic neurons loss induced by 192-IgG-SAP, it avoided the neuronal damage progression.


Assuntos
Transtornos da Memória/tratamento farmacológico , Molsidomina/farmacologia , Reconhecimento Psicológico/efeitos dos fármacos , Acetilcolina/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Colina O-Acetiltransferase/metabolismo , Neurônios Colinérgicos/efeitos dos fármacos , Cognição/efeitos dos fármacos , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Hipocampo/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Memória de Curto Prazo/fisiologia , Molsidomina/metabolismo , Doadores de Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Ratos , Ratos Wistar , Saporinas/farmacologia , Percepção Visual/efeitos dos fármacos
8.
Front Behav Neurosci ; 12: 202, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30233338

RESUMO

The effects caused by exposure to lead (Pb) are still considered as a relevant health risk despite public policies aimed to restricting the use of this element. The toxicity limit in the blood (10 µg/dL, established by the Center for Disease Control and Prevention) has been insufficient to prevent adverse effects and even lower values have been related to neurobehavioral dysfunctions in children. Currently, there is not a safe limit of exposure to Pb. A large body of evidence points to environmental pollutant exposure as the cause of predisposition to violent behavior, among others. Considering the evidence by our group and others, we propose that Pb exposure induces alterations in the brain vasculature, specifically in nitric oxide synthases (NOS), affecting in turn the serotonergic system and leading to heightened aggressive behavior in the exposed individuals. This review article describes the consequences of Pb exposure on the nitrergic and serotonergic systems as well as its relationship with aggressive behavior. In addition, it summarizes the available therapy to prevent damage in gestation and among infants.

10.
Nutr Neurosci ; 20(7): 388-395, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26928375

RESUMO

Essential fatty acids have an important effect on oxidative stress-related diseases. The Huntington's disease (HD) is a hereditary neurologic disorder in which oxidative stress caused by free radicals is an important damage mechanism. The HD experimental model induced by quinolinic acid (QUIN) has been widely used to evaluate therapeutic effects of antioxidant compounds. The aim of this study was to test whether the fatty acid content in olive- or fish-oil-rich diet prevents against QUIN-related oxidative damage in rats. Rats were fed during 20 days with an olive- or a fish-oil-rich diet (15% w/w). Posterior to diet period, rats were striatally microinjected with QUIN (240 nmol/µl) or saline solution. Then, we evaluated the neurological damage, oxidative status, and gamma isoform of the peroxisome proliferator-activated receptor (PPARγ) expression. Results showed that fatty acid-rich diet, mainly by fish oil, reduced circling behavior, prevented the fall in GABA levels, increased PPARγ expression, and prevented oxidative damage in striatal tissue. In addition none of the enriched diets exerted changes neither on triglycerides or cholesterol blood levels, nor or hepatic function. This study suggests that olive- and fish-oil-rich diets exert neuroprotective effects.


Assuntos
Corpo Estriado/efeitos dos fármacos , Ácidos Graxos Essenciais/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ácido Quinolínico/toxicidade , Animais , Peso Corporal , Colesterol/sangue , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Óleos de Peixe/farmacologia , Doença de Huntington/tratamento farmacológico , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Fármacos Neuroprotetores/farmacologia , Azeite de Oliva/farmacologia , Ratos , Ratos Wistar , Triglicerídeos/sangue , Ácido gama-Aminobutírico/metabolismo
11.
J Neurosci Res ; 95(7): 1495-1502, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27753128

RESUMO

Lesions of the cerebellar dentate nucleus (DN) reduce the after-discharge duration induced by repetitive kindling stimulation and decrease seizures to a lower rank according to Racine's scale. The DN sends cholinergic and glutamatergic fibers to the red nucleus (RN), which is composed of glutamatergic and GABAergic cells. To test the participation of these neurotransmitters in seizures, we compared the levels of glutamate and gamma-aminobutyric acid (GABA) at the RN in a control condition, a kindled stage, and a kindled stage followed by DN lesions. We found that the kindled stage was associated with significant reductions in glutamate and GABA in the RN and that the lesions of the DN in kindled rats reversed the severity of seizures and restored the GABA levels. GAD65 , a GABA-synthesizing enzyme, was increased in kindled rats and decreased after DN lesions. GAD65 commonly appears localized at nerve terminals and synapses, and it is only activated when GABA neurotransmission occurs. Thus, it is possible that the increased expression of GAD65 found in kindled rats could be due to an exacerbated demand for GABA due to kindled seizures. It is known that GABA maintains the inhibitory tone that counterbalances neuronal excitation. The decreased expression of GAD65 found after the DN lesions indicated that the GABA-synthesizing enzyme was no longer required once it eliminated the excitatory glutamate input to the RN. We thus conclude that DN lesions and their consequent biochemical changes are capable of decreasing the generalized seizures induced by kindling stimulation. © 2016 Wiley Periodicals, Inc.


Assuntos
Giro Denteado/metabolismo , Modelos Animais de Doenças , Epilepsia/metabolismo , Excitação Neurológica/fisiologia , Núcleo Rubro/metabolismo , Tonsila do Cerebelo/anatomia & histologia , Tonsila do Cerebelo/metabolismo , Animais , Giro Denteado/anatomia & histologia , Epilepsia/patologia , Ácido Glutâmico/metabolismo , Locomoção/fisiologia , Masculino , Vias Neurais/metabolismo , Vias Neurais/patologia , Ratos , Ratos Wistar , Núcleo Rubro/anatomia & histologia , Ácido gama-Aminobutírico/metabolismo
12.
Biol Trace Elem Res ; 162(1-3): 211-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25319005

RESUMO

Quinolinic acid (QUIN) striatal injection in rat reproduces the main neurochemical features of Huntington's disease (HD), including oxidative damage. In this study, we evaluated the effect of a copper (Cu) supplement in drinking water (90 ppm Cu, 28 days) on the QUIN-induced HD model in the rat. Copper exposure caused no signs of liver toxicity; however, it produced significant Cu accumulation in striatum. It is noteworthy that QUIN also caused increased striatal Cu content; when the supplement was administered to animals with QUIN-injury, an even higher metal striatal accumulation was observed. Cu pre-treatment preserved striatal gamma-aminobutyric acid (GABA) content, which was reduced by QUIN intrastriatal injection. Similarly, apomorphine-induced circling behavior was reduced in Cu-pretreated QUIN-damaged rats. Metal supplement in drinking water prevented both lipid peroxidation and reactive oxygen species (ROS) formation caused by QUIN in striatum. In Cu-treated groups, superoxide dismutase-1 (SOD1) activity showed a significant increase, while SOD2 activity was slightly enhanced. Although the pathophysiological role for higher Cu levels in patients with HD and in experimental models of the disease is not fully understood, results in the present study suggest that Cu oral intake stimulates anti-oxidant defenses, an effect that may be a potential factor for reducing the progression of HD.


Assuntos
Cobre/uso terapêutico , Doença de Huntington/tratamento farmacológico , Doença de Huntington/metabolismo , Animais , Apomorfina/toxicidade , Cobre/farmacologia , Modelos Animais de Doenças , Doença de Huntington/induzido quimicamente , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ácido Quinolínico/toxicidade , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Ácido gama-Aminobutírico/metabolismo
13.
Inhal Toxicol ; 26(8): 485-91, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24987980

RESUMO

The World Health Organization identified urban outdoor air pollution as the eighth highest mortality risk factor in high-income countries. Exposure to ambient pollutants such as ozone (O3) increases the number of hospital admissions. O3 is a highly reactive gas that reacts with cells lining the airways, producing the formation of reactive oxygen species and inflammation. Beyond the respiratory system, O3 exposure also produces fatigue, lethargy, headaches, and significant decrease in rapid-eye-movement sleep related to an increase in slow-wave sleep. Interestingly, these sleep changes can be significantly mitigated by treatment with indomethacin, which suggests that an inflammatory mechanism may be responsible for these neurological symptoms. To characterize the inflammatory mechanisms by which O3 affects tissues outside the pulmonary system, we evaluated inflammatory factors in both lung and brain. Rats exposed to 1 part per million O3 for 1, 3 or 6 h, as well as rats exposed daily for 1 or 3 h over five consecutive days, showed increases in TNF-α and IL-6 levels within the lungs as well as increases in TNF-α, IL-6, NF-κB p50 and GFAP levels in the cerebral cortex. These results support the hypothesis that the neuroinflammatory response may be responsible for the central nervous system effects of O3 exposure.


Assuntos
Poluentes Atmosféricos/toxicidade , Córtex Cerebral/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Oxidantes/toxicidade , Ozônio/toxicidade , Animais , Córtex Cerebral/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Inflamação/induzido quimicamente , Interleucina-6/metabolismo , Pulmão/metabolismo , Masculino , NF-kappa B/metabolismo , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo
14.
Rev Neurosci ; 24(3): 337-52, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23585211

RESUMO

Ozone (O3) is a component of photochemical smog, which is a major air pollutant and demonstrates properties that are harmful to health because of the toxic properties that are inherent to its powerful oxidizing capabilities. Environmental O3 exposure is associated with many symptoms related to respiratory disorders, which include loss of lung function, exacerbation of asthma, airway damage, and lung inflammation. The effects of O3 are not restricted to the respiratory system or function - adverse effects within the central nervous system (CNS) such as decreased cognitive response, decrease in motor activity, headaches, disturbances in the sleep-wake cycle, neuronal dysfunctions, cell degeneration, and neurochemical alterations have also been described; furthermore, it has also been proposed that O3 could have epigenetic effects. O3 exposure induces the reactive chemical species in the lungs, but the short half-life of these chemical species has led some authors to attribute the injurious mechanisms observed within the lungs to inflammatory processes. However, the damage to the CNS induced by O3 exposure is not well understood. In this review, the basic mechanisms of inflammation and activation of the immune system by O3 exposure are described and the potential mechanisms of damage, which include neuroinflammation and oxidative stress, and the signs and symptoms of disturbances within the CNS caused by environmental O3 exposure are discussed.


Assuntos
Sistema Nervoso/efeitos dos fármacos , Oxidantes Fotoquímicos/toxicidade , Ozônio/toxicidade , Poluentes Atmosféricos/toxicidade , Animais , Exposição Ambiental , Humanos , Sistema Nervoso/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/metabolismo
15.
Cancer Chemother Pharmacol ; 69(2): 495-504, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21861129

RESUMO

Resistance and toxicity are the major barriers to successful cancer chemotherapies. Developing molecules that reduce drug resistance and improve antineoplastic effects is of great interest for cancer research; ideally, these substances should not affect the pharmacodynamics of the chemotherapeutic agent while providing a synergistic antineoplastic effect. In this study, we tested in vitro co-administration of the antineoplastic agents cisplatin or paclitaxel with probenecid, an anion channel inhibitor, in a panel of cancer cell lines to determine the cytotoxicity and synergistic effects of these drug combinations. In addition, we measured the clonogenicity and apoptotic index in these cells. We observed a synergistic interaction between probenecid and the chemotherapeutic agents, and increasing doses of probenecid resulted in a significant decrease in the effective doses of the chemotherapeutic agents. For the antineoplastic agent and probenecid combinations, we found increased cell death, reduced colony formation, and a higher number of apoptotic cells, compared with treatment of cisplatin or paclitaxel alone. Further research is necessary to elucidate the molecular mechanisms by which the synergistic effect occurs. If these synergistic effects can be reproduced in vivo, the co-administration of probenecid with different chemotherapeutic agents may provide a valid treatment in patients with chemotherapy resistance.


Assuntos
Apoptose/efeitos dos fármacos , Cisplatino/farmacologia , Paclitaxel/farmacologia , Probenecid/farmacologia , Adjuvantes Farmacêuticos/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Células HeLa , Humanos , Microscopia de Fluorescência , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Ensaio Tumoral de Célula-Tronco
16.
Rev Invest Clin ; 61(5): 405-11, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20184101

RESUMO

INTRODUCTION: Parkinson's disease (PD) is a neurodegenerative disorder characterized by the presence of motor disturbances, derived from the striatal dopamine depletion. Previously, we reported that CuSO4 pretreatment blocked an oxidative stress marker (lipid peroxidation) and prevented the striatal dopamine depletion induced by the administration of the 1-methyl-4-phenylpiridinium (MPP+), the toxic metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a model of PD. OBJECTIVE: . To determine if tyrosine hydroxylase (TH), the rate-limiting synthetic enzyme of dopamine, is implicated in the neuroprotective effect of CuSO4 pretreatment, and if this neuroprotective effect is able to prevent the hypokinetic state (measured as spontaneous locomotor activity, SLA) induced by the experimental model of PD. MATERIAL AND METHODS: C57 Black/6J mice received a single dose of CuSO4 (2.5 mg/kg, i.p.) either 16 or 24 h before the administration of MPP+ (18 microg/3 microl, i.c.v.). Twenty four hours later, mice SLA was registered and animals sacrificed. Striatal L-DOPA accumulation derived from the administration of a central dopamine descarboxilase inhibitor was evaluated, a strategy considered as a reliable indirect analysis of tyrosine hydroxylase activity (THA). RESULTS: Administration of, MPP+ decreased SLA (-52%; p = 0.003) as compared to control group values, whereas those mice pretreated with CuSO4 16 h before MPP+, increased SLA by 47% as compared with control group (p = 0.015). Mice pretreated with CuSO4 24 h before MPP+, also showed a statistically significant increase in SLA (71%; p = 0.02), when compared with control group. As a consequence of MPP+ administration, THA was also reduced as compared to control group values (32%; p < 0.05). Reduction of THA was blocked when mice were pretreated with CuSO4 16 h before MPP+. Moreover, mice receiving the CuSO4 24 h before MPP+ showed a significant increase (38%; p < 0.05) in THA when compared with control group. CONCLUSION: Results suggest that preservation of THA participates in the neuroprotective effects derived from the copper supplementation, a phenomenon that avoid the hypokinetic state induced by the MPP+ experimental model of PD.


Assuntos
Sulfato de Cobre/uso terapêutico , Modelos Animais de Doenças , Doença de Parkinson/tratamento farmacológico , Tirosina 3-Mono-Oxigenase/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doença de Parkinson/enzimologia , Doença de Parkinson/fisiopatologia
17.
J Neurosci Res ; 85(7): 1391-402, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17387708

RESUMO

Nitric oxide (NO) is an essential messenger molecule in brain, where it is produced in neurons mostly by the activity of the neuronal isoform of nitric oxide synthase (nNOS). To understand the participation of the different isoforms of NOS in physiological functioning and in pathological processes, mice with null mutations for each of the NOS isoforms have been generated. In the present paper, we report that there is a selective protection from oxidative damage in the brain of mice with a targeted disruption of the nNOS gene. The cerebellum of these mice shows reduced levels of lipid peroxidation (LP) at the different ages tested, compared with wild-type mice, and also a reduction in the formation of reactive oxygen species (ROS). We observed a decrease of LP in cortex, and no effect on either LP or ROS formation was observed in striatum of knockout mice compared with wild type. We also report increased spontaneous motor activity of knockout mice. The expression and activity of nNOS are crucial to maintain redox status in brain, and we consider that the alteration in oxidative damage may help us to explain the phenotypical characteristics of nNOS knockout mice and their differential susceptibility to brain insults.


Assuntos
Encéfalo/enzimologia , Peroxidação de Lipídeos/fisiologia , Óxido Nítrico Sintase Tipo I/metabolismo , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Fatores Etários , Análise de Variância , Animais , Cerebelo/enzimologia , Córtex Cerebral/enzimologia , Distribuição de Qui-Quadrado , Isoenzimas , Masculino , Camundongos , Camundongos Knockout , Neostriado/enzimologia , Óxido Nítrico Sintase Tipo I/genética , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...