Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Anticancer Drugs ; 32(2): 117-126, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33136700

RESUMO

In this study, we investigated the putative cytotoxic effect elicited by the garlic-derived compound S-allylcysteine (SAC) in two human cancer cell lines (HCC827 and NCI-H1975) in order to develop an experimental approach to the therapeutic potential of this molecule for lung cancer. Cells were incubated for 24, 48 and 72 h in the presence of SAC (10 or 20 mM), which resulted in a concentration- and time-dependent decrease in cell viability and culture confluence in both cell lines. These effects were contrasted with - and validated through - those observed in an immortalized but nontumorigenic epithelial cell line from human bronchial epithelium (BEAS-2B, negative control) and an adenocarcinoma human alveolar basal epithelial cell line (A549, positive control). SAC (20 mM at 72 h) also increased the oxidative damage to lipids, augmented apoptosis, and decreased the expression of the nuclear factor erythroid 2-related factor 2 (Nrf2) and the nuclear factor kappa B (NF-κB) proteins in HCC827 and NCI-H1975 cells. Our results establish the efficacy of SAC in reducing malignant growth and proliferation of lung tumor cells. This effect is mediated by the induction of oxidative damage associated with the downregulation of Nrf2 and NF-κB and their corresponding signaling pathways.


Assuntos
Antineoplásicos/farmacologia , Cisteína/análogos & derivados , Neoplasias Pulmonares/tratamento farmacológico , Fator 2 Relacionado a NF-E2/biossíntese , NF-kappa B/biossíntese , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cisteína/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais , Fatores de Tempo
2.
Cell Tissue Bank ; 19(4): 569-580, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30155658

RESUMO

In this work we estimated the budgetary impact of the samples produced by the biobank of the "Instituto Nacional de Cancerología" (BT-INCan) to set a recuperation fee from the perspective of the Health Ministry of Mexico. The study is an observational retrospective review of the direct medical costs (DMCs) of the processes involved in cryopreservation of the samples collected, on a per sample basis, including materials, laboratory tests, personnel, and administrative costs. Materials and labor costs were determined by information collected from the BT-INCan. DMCs were provided depending on the type of sample: plasma, tissue and biopsy; they were calculated according to the process required to preserve them. Sensitivity analysis was performed using bootstrap. Recuperation costs ranged from 130 to 155 USD. Costs were considered on a 5-year time frame for the maintenance per sample, which is the average time that a sample is kept in the BT-INCan. The cost analysis is perceived as an approximation to the most adequate recuperation fee per sample needed to guarantee the correct development of the BT-INCan. This work provides a basis and valuable information about costs, to enable several health institutions to strategically plan and manage a biobank or even motivate to establish their own biobank.


Assuntos
Bancos de Espécimes Biológicos/economia , Farmacoeconomia , Preservação Biológica/economia , Custos e Análise de Custo , Humanos , México
3.
Neurochem Res ; 41(10): 2619-2626, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27278758

RESUMO

3-Methylglutaric acid (3MGA) is an organic acid that accumulates in various organic acidemias whose patients present neurodegeneration events in children coursing with metabolic acidurias. Limited evidence describes the toxic mechanisms elicited by 3MGA in the brain. Herein, we explored the effects of 3MGA on different toxic endpoints in synaptosomal and mitochondrial-enriched fractions of adult rat brains to provide novel information on early mechanisms evoked by this metabolite. At 1 and 5 mM concentration, 3MGA increased lipid peroxidation, but decreased mitochondrial function only at 5 mM concentration. Despite less intense effects were obtained at 1 mM concentration, its co-administration with the kynurenine pathway (KP) metabolite and N-methyl-D-aspartate receptor (NMDAr) agonist, quinolinic acid (QUIN, 50 and 100 µM), produced toxic synergism on markers of oxidative stress and mitochondrial function. The toxicity of 3MGA per se (5 mM) was prevented by the cannabinoid receptor agonist WIN55,212-2 and the NMDAr antagonist kynurenic acid (KYNA), suggesting cannabinoid and glutamatergic components in the 3MGA pattern of toxicity. The synergic model (3MGA + QUIN) was also sensitive to KYNA and the antioxidant S-allylcysteine, but not to the nitric oxide synthase inhibitor L-nitroarginine methyl ester. These findings suggest various underlying mechanisms involved in the neurotoxicity of 3MGA that may possibly contribute to the neurodegeneration observed in acidemias.


Assuntos
Encéfalo/efeitos dos fármacos , Meglutol/análogos & derivados , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Sinaptossomos/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Encéfalo/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Meglutol/farmacologia , Mitocôndrias/metabolismo , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Receptores de Canabinoides/metabolismo , Sinaptossomos/metabolismo
4.
Neurotox Res ; 41(6): 514-525, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37458923

RESUMO

Inhibition of enzymes responsible for endocannabinoid hydrolysis represents an invaluable emerging tool for the potential treatment of neurodegenerative disorders. Monoacylglycerol lipase (MAGL) is the enzyme responsible for degrading 2-arachydonoylglycerol (2-AG), the most abundant endocannabinoid in the central nervous system (CNS). Here, we tested the effects of the selective MAGL inhibitor JZL184 on the 3-nitropropinic acid (3-NP)-induced short-term loss of mitochondrial reductive capacity/viability and oxidative damage in rat brain synaptosomal/mitochondrial fractions and cortical slices. In synaptosomes, while 3-NP decreased mitochondrial function and increased lipid peroxidation, JZL184 attenuated both markers. The protective effects evoked by JZL184 on the 3-NP-induced mitochondrial dysfunction were primarily mediated by activation of cannabinoid receptor 2 (CB2R), as evidenced by their inhibition by the selective CB2R inverse agonist JTE907. The cannabinoid receptor 1 (CB1R) also participated in this effect in a lesser extent, as evidenced by the CB1R antagonist/inverse agonist AM281. In contrast, activation of CB1R, but not CB2R, was responsible for the protective effects of JZL184 on the 3-NP-iduced lipid peroxidation. Protective effects of JZL184 were confirmed in other toxic models involving excitotoxicity and oxidative damage as internal controls. In cortical slices, JZL184 ameliorated the 3-NP-induced loss of mitochondrial function, the increase in lipid peroxidation, and the inhibition of succinate dehydrogenase (mitochondrial complex II) activity, and these effects were independent on CB1R and CB2R, as evidenced by the lack of effects of AM281 and JTE907, respectively. Our novel results provide experimental evidence that the differential protective effects exerted by JZL184 on the early toxic effects induced by 3-NP in brain synaptosomes and cortical slices involve MAGL inhibition, and possibly the subsequent accumulation of 2-AG. These effects involve pro-energetic and redox modulatory mechanisms that may be either dependent or independent of cannabinoid receptors' activation.


Assuntos
Endocanabinoides , Sinaptossomos , Ratos , Animais , Sinaptossomos/metabolismo , Monoacilglicerol Lipases/metabolismo , Receptores de Canabinoides , Agonismo Inverso de Drogas , Encéfalo/metabolismo , Estresse Oxidativo , Benzodioxóis/farmacologia , Receptor CB1 de Canabinoide
5.
Pharmaceuticals (Basel) ; 15(7)2022 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-35890085

RESUMO

Metformin has been under basic and clinical study as an oncological repurposing pharmacological agent for several years, stemming from observational studies which consistently evidenced that subjects who were treated with metformin had a reduced risk for development of cancer throughout their lives, as well as improved survival outcomes when diagnosed with neoplastic diseases. As a result, several basic science studies have attempted to dissect the relationship between metformin's metabolic mechanism of action and antineoplastic cellular signaling pathways. Evidence in this regard was compelling enough that a myriad of randomized clinical trials was planned and conducted in order to establish the effect of metformin treatment for patients with diverse neoplasms, including lung cancer. As with most novel antineoplastic agents, early results from these studies have been mostly discouraging, though a recent analysis that incorporated body mass index may provide significant information regarding which patient subgroups might derive the most benefit from the addition of metformin to their anticancer treatment. Much in line with the current pipeline for anticancer agents, it appears that the benefit of metformin may be circumscribed to a specific patient subgroup. If so, addition of metformin to antineoplastic agents could prove one of the most cost-effective interventions proposed in the context of precision oncology. Currently published reviews mostly rely on a widely questioned mechanism of action by metformin, which fails to consider the differential effects of the drug in lean vs. obese subjects. In this review, we analyze the pre-clinical and clinical information available to date regarding the use of metformin in various subtypes of lung cancer and, further, we present evidence as to the differential metabolic effects of metformin in lean and obese subjects where, paradoxically, the obese subjects have reported more benefit with the addition of metformin treatment. The novel mechanisms of action described for this biguanide may explain the different results observed in clinical trials published in the last decade. Lastly, we present novel hypothesis regarding potential biomarkers to identify who might reap benefit from this intervention, including the role of prolyl hydroxylase domain 3 (PHD3) expression to modify metabolic phenotypes in malignant diseases.

6.
Plant Foods Hum Nutr ; 66(4): 348-54, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21850441

RESUMO

Different garlic products reduce the cerebral ischemic damage due to their antioxidant properties. In this work, we investigated the effect of aged garlic extract (AGE) on cyclooxygenase-2 (COX-2) protein levels and activity, and its role as a possible mechanism of neuroprotection in a cerebral ischemia model. Animals were subjected to 1 h of ischemia plus 24 h of reperfusion. AGE (1.2 ml/kg weight, i.p.) was administered at onset of reperfusion. To evaluate the damage induced by cerebral ischemia, the neurological deficit, the infarct area, and the histological alterations were measured. As an oxidative stress marker to deoxyribonucleic acid, 8-hydroxy-2-deoxyguanosine (8-OHdG) levels were determined. Finally, as inflammatory markers, TNFα levels and COX-2 protein levels and activity were measured. AGE treatment diminished the neurological alterations (61.6%), the infarct area (54.8%) and the histological damage (37.7%) induced by cerebral ischemia. AGE administration attenuated the increase in 8-OHdG levels (77.8%), in TNFα levels (76.6%), and in COX-2 protein levels (73.6%) and activity (30.7%) induced after 1 h of ischemia plus 24 h of reperfusion. These data suggest that the neuroprotective effect of AGE is associated not only to its antioxidant properties, but also with its capacity to diminish the increase in TNFα levels and COX-2 protein expression and activity. AGE may have the potential to attenuate the cerebral ischemia-induced inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Isquemia Encefálica/complicações , Infarto Cerebral/prevenção & controle , Ciclo-Oxigenase 2/metabolismo , Alho/química , Extratos Vegetais/farmacologia , 8-Hidroxi-2'-Desoxiguanosina , Animais , Anti-Inflamatórios/química , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Isquemia Encefálica/induzido quimicamente , Infarto Cerebral/etiologia , Ciclo-Oxigenase 2/genética , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análise , Desoxiguanosina/metabolismo , Masculino , Modelos Animais , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/química , Distribuição Aleatória , Ratos , Ratos Wistar , Reperfusão/efeitos adversos , Fatores de Tempo , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/metabolismo
7.
Neurotox Res ; 39(2): 146-155, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33141426

RESUMO

Endocannabinoid-based therapies constitute an emerging tool for the potential treatment of neurodegenerative disorders, requiring characterization at the experimental level. The effects of URB597, an inhibitor of the fatty acid amide hydrolase (FAAH), were tested against the quinolinic acid (QUIN)-induced early toxic effects in rat cortical slices, and compared with those effects exerted by the endocannabinoid anandamide (AEA). URB597 prevented the QUIN-induced loss of mitochondrial function/cell viability and lipid peroxidation, while reduced necrosis, and to a lesser extent, apoptosis. The protective effects of URB597 were mediated by activation of cannabinoid receptor 1 (CB1r), as evidenced by their inhibition by the selective CB1r antagonist AM281. Similar effects were observed when testing AEA against QUIN toxicity. Our findings demonstrate the neuroprotective properties of URB597 during the early stages of excitotoxic damage to cortical tissue, suggesting that these properties are mediated by FAAH inhibition, and might be linked to the protective effects of AEA, or the combination of endocannabinoids.


Assuntos
Amidoidrolases/antagonistas & inibidores , Benzamidas/administração & dosagem , Carbamatos/administração & dosagem , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Ácido Quinolínico/toxicidade , Receptor CB1 de Canabinoide/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Masculino , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos Wistar
8.
Neurotox Res ; 39(6): 2072-2097, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34741755

RESUMO

The endocannabinoid system (ECS) is composed of endogenous cannabinoids; components involved in their synthesis, transport, and degradation; and an expansive variety of cannabinoid receptors. Hypofunction or deregulation of the ECS is related to pathological conditions. Consequently, endogenous enhancement of endocannabinoid levels and/or regulation of their metabolism represent promising therapeutic approaches. Several major strategies have been suggested for the modulation of the ECS: (1) blocking endocannabinoids degradation, (2) inhibition of endocannabinoid cellular uptake, and (3) pharmacological modulation of cannabinoid receptors as potential therapeutic targets. Here, we focused in this review on degradation/reuptake inhibitors over cannabinoid receptor modulators in order to provide an updated synopsis of contemporary evidence advancing mechanisms of endocannabinoids as pharmacological tools with therapeutic properties for the treatment of several disorders. For this purpose, we revisited the available literature and reported the latest advances regarding the biomedical properties of fatty acid amide hydrolase and monoacylglycerol lipase inhibitors in pre-clinical and clinical studies. We also highlighted anandamide and 2-arachidonoylglycerol reuptake inhibitors with promising results in pre-clinical studies using in vitro and animal models as an outlook for future research in clinical trials.


Assuntos
Endocanabinoides/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Endocanabinoides/antagonistas & inibidores , Endocanabinoides/fisiologia , Humanos
9.
J Immunol Res ; 2021: 6632249, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34195295

RESUMO

BACKGROUND: Lung neuroendocrine tumors account for approximately 15% of all lung cancer cases. LNET are subdivided into typical carcinoid (TC), atypical carcinoid (AC), large cell neuroendocrine carcinoma (LCNEC), and small-cell lung cancer (SCLC). The Ki-67 index has been used for decades to evaluate mitotic counts however, the role of Ki-67 as a biomarker for assessing prognosis and guiding therapy in metastatic LNET still lacks feasible clinical validation. Recent clinical trials have indicated that inhibition of CD47 with anti-CD47 antibodies exerts a promising antitumor effect against several human malignancies, including NSCLC, melanoma, and hematologic malignancies. However, the clinical relevance of CD47 expression in LNET has remained unclear. METHODS: We performed a retrospective study in which we analyzed tumor biopsies from 51 patients with a confirmed diagnosis of LNET that received treatment at our hospital. Then, we analyzed if there was any correlation between CD47 expression with any clinical or pathological characteristic. We also analyzed the prognostic significance of CD47, assessed as progression-free survival and overall survival. RESULTS: A total of 51 patients with LNET were enrolled in our study. The mean age at diagnosis was 57.6 (±11.6) years; 30 patients were women (59%). 27.5% of patients were positive for CD47 expression, and 72.5% of patients showed a CD47 expression of less than 1% and were considered as negatives. In patients with high-grade tumors (this time defined as Ki-67 > 40%), the positive expression of CD47 was strongly associated with an increased PFS. Albeit, these differences did not reach statistical significance when analyzing OS. CONCLUSION: Contrary to what happens in a wide range of hematologic and solid tumors, a higher expression of CD47 in patients with LNET is associated with a better progression-free survival, especially in patients with a Ki-67 ≥ 40%. This "paradox" remains to be confirmed and explained by larger studies.


Assuntos
Biomarcadores Tumorais/metabolismo , Antígeno CD47/metabolismo , Neoplasias Pulmonares/metabolismo , Pulmão/patologia , Tumores Neuroendócrinos/metabolismo , Idoso , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/diagnóstico , Tumores Neuroendócrinos/mortalidade , Prognóstico , Análise de Sobrevida , Regulação para Cima
10.
J Med Food ; 24(10): 1068-1075, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33872073

RESUMO

Acrocomia aculeata fruits are rich in monounsaturated fatty acid, ß-carotene, tocopherol, and other antioxidant compounds. The aim of our study was to investigate and compare the protective effects of A. aculeata pulp oil and microencapsulated pulp oil on brain oxidative damage induced by chronic restraint stress (CRS) in rats (cortex, hippocampus, and striatum). Thirty-six Wistar rats were divided into six treatment groups: C, P, and M groups received 1 µL/g of body weight of distilled water, pulp oil, and pulp oil microcapsules by daily gavage, respectively. The SC, SP, and SM groups received 1 µL/g of body weight of distilled water, pulp oil, and pulp oil microcapsules by daily gavage, respectively, and were then subjected to uninterrupted 6 h of CRS. After 21 days of testing, the rats were euthanized and the brain tissue of the groups was removed for evaluation for oxidative damage markers and antioxidant enzymes. Endpoints of oxidative stress (OS) markers (lipid peroxidation, protein carbonylation, and reduced glutathione [GSH]) and antioxidant enzymes (superoxide dismutase and catalase) were evaluated. By imposing chronic stress on rats, pulp oil and microcapsules of pulp oil induced positive antioxidant responses, mainly by increasing the GSH content, increasing the ability of neural tissues to deal with inherent OS, thus protecting against neurodegenerative diseases. The administration of A. aculeata pulp oil and microencapsulated pulp oil made the reversal of the oxidant parameters, which may protect the brain tissue of rats altered by CRS. The Clinical Trial Registration number: n° 1.008/2018 CEUA/UFMS.


Assuntos
Arecaceae , Fármacos Neuroprotetores , Animais , Antioxidantes , Cápsulas , Ratos , Ratos Wistar
11.
Neuroscience ; 443: 206-217, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32681927

RESUMO

Neuroprotective action of the novel aldose reductase (AR) inhibitor cemtirestat (CMT), 2-(3-thioxo-2H-[1,2,4]triazino[5,6-b]indol-5(3H)-yl)acetic acid, was recently proved in experimental rat models of diabetes. The in vivo results indicated that the antioxidant activity of this compound might have participated on its effects. The aim of this study was to explore in a greater detail the putative antioxidant mechanisms potentially involved in CMT mediated neuroprotection. Antioxidant efficacy per se of CMT was proved by a ferric reducing antioxidant power (FRAP) test and CMT was found to scavenge reactive oxygen species (ROS) generated in water phase chemically with decreasing efficacy as follows ROO > H2O2 > O2-. Studies in liposomes revealed the ability of CMT to inhibit lipid peroxidation more efficiently than melatonin, yet less effectively than Trolox. In the rat brain cortical slices, CMT reduced the loss of cell viability/mitochondrial function induced by quinolinic acid (QUIN), and inhibited lipid peroxidation. In addition, CMT normalized the GSH/GSSG ratio which could be explained, at least partially, by the ability of this compound to release free GSH from the pool of endogenously bound disulfides. Neuronal cell damage induced by QUIN or H2O2 was reduced by CMT as proved by significant drop in propidium iodide incorporation into cells. On balance then, our results corroborated the notion of a multifunctional action of CMT as a drug combining AR inhibition with direct antioxidant and ROS scavenging activity. Moreover, the ability of CMT to restore thiol-disulfide homeostasis was proved.


Assuntos
Antioxidantes , Lipossomos , Animais , Antioxidantes/farmacologia , Encéfalo , Peróxido de Hidrogênio , Ácidos Indolacéticos , Peroxidação de Lipídeos , Modelos Químicos , Neuroproteção , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio , Compostos de Sulfidrila
12.
Neurotox Res ; 38(4): 929-940, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32813208

RESUMO

Neuroprotective approaches comprising different mechanisms to counteract the noxious effects of excitotoxicity and oxidative stress need validation and detailed characterization. Although S-allylcysteine (SAC) is a natural compound exhibiting a broad spectrum of protective effects characterized by antioxidant, anti-inflammatory, and neuromodulatory actions, the mechanisms underlying its protective role on neuronal cell damage triggered by early excitotoxic insults remain elusive. In this study, we evaluated if the preconditioning or the post-treatment of isolated rat cortical slices with SAC (100 µM) can ameliorate the toxic effects induced by the excitotoxic metabolite quinolinic acid (QUIN, 100 µM), and whether this protective response involves the early display of specific antioxidant and neuroprotective signals. For this purpose, cell viability/mitochondrial reductive capacity, lipid peroxidation, levels of reduced and oxidized glutathione (GSH and GSSG, respectively), the rate of cell damage, the NF-E2-related factor 2/antioxidant response element (Nrf2/ARE) binding activity, heme oxygenase 1 (HO-1) regulation, extracellular signal-regulated kinase (ERK1/2) phosphorylation, and the levels of tumor necrosis factor-alpha (TNF-α) and the neurotrophin brain-derived neurotrophic factor (BDNF) were all estimated in tissue slices exposed to SAC and/or QUIN. The incubation of slices with QUIN augmented all toxic endpoints, whereas the addition of SAC prevented and/or recovered all toxic effects of QUIN, exhibiting better results when administered 60 min before the toxin and demonstrating protective and antioxidant properties. The early stimulation of Nrf2/ARE binding activity, the upregulation of HO-1, the ERK1/2 phosphorylation and the preservation of BDNF tissue levels by SAC demonstrate that this molecule displays a wide range of early protective signals by triggering orchestrated antioxidant responses and neuroprotective strategies. The relevance of the characterization of these mechanisms lies in the confirmation that the protective potential exerted by SAC begins at the early stages of excitotoxicity and neurodegeneration and supports the design of integral prophylactic/therapeutic strategies to reduce the deleterious effects observed in neurodegenerative disorders with inherent excitotoxic events.


Assuntos
Elementos de Resposta Antioxidante/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Córtex Cerebral/metabolismo , Cisteína/análogos & derivados , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Elementos de Resposta Antioxidante/fisiologia , Córtex Cerebral/efeitos dos fármacos , Cisteína/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/fisiologia , Masculino , Fármacos Neuroprotetores/farmacologia , Técnicas de Cultura de Órgãos , Estresse Oxidativo/fisiologia , Ligação Proteica/fisiologia , Ratos , Ratos Wistar
13.
Toxins (Basel) ; 11(2)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30717279

RESUMO

Zoanthids of the genus Palythoa are distributed worldwide in shallow waters around coral reefs. Like all cnidarians, they possess nematocysts that contain a large diversity of toxins that paralyze their prey. This work was aimed at isolating and functionally characterizing a cnidarian neurotoxic phospholipase named A2-PLTX-Pcb1a for the first time. This phospholipase was isolated from the venomous extract of the zoanthid Palythoa caribaeorum. This enzyme, which is Ca2+-dependent, is a 149 amino acid residue protein. The analysis of the A2-PLTX-Pcb1a sequence showed neurotoxic domain similitude with other neurotoxic sPLA2´s, but a different catalytic histidine domain. This is remarkable, since A2-PLTX-Pcb1a displays properties like those of other known PLA2 enzymes.


Assuntos
Antozoários , Córtex Motor/efeitos dos fármacos , Síndromes Neurotóxicas , Neurotoxinas/toxicidade , Fosfolipases A2/toxicidade , Animais , Comportamento Animal/efeitos dos fármacos , Injeções Intraventriculares , Masculino , Atividade Motora/efeitos dos fármacos , Córtex Motor/patologia , Neurotoxinas/química , Neurotoxinas/isolamento & purificação , Fosfolipases A2/química , Fosfolipases A2/isolamento & purificação , Ratos Wistar
14.
Antioxid Redox Signal ; 28(18): 1626-1651, 2018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-28467722

RESUMO

SIGNIFICANCE: Production of pro-inflammatory and anti-inflammatory cytokines is part of the defense system that mostly microglia and macrophages display to induce normal signaling to counteract the deleterious actions of invading pathogens in the brain. Also, redox activity in the central nervous system (CNS) constitutes an integral part of the metabolic processes needed by cells to exert their normal molecular and biochemical functions. Under normal conditions, the formation of reactive oxygen and nitrogen species, and the following oxidative activity encounter a healthy balance with immunological responses to preserve cell functions in the brain. However, under different pathological conditions, inflammatory responses recruit pro-oxidant signals and vice versa. The aim of this article is to review the basic concepts about the triggering of inflammatory and oxidative responses in the CNS. Recent Advances: Diverse concurrent toxic pathways are described to provide a solid mechanistic scope for considering intervention at the experimental and clinical levels that are aimed at diminishing the harmful actions of these two contributing factors to nerve cell damage. Critical Issues and Future Directions: The main conclusion supports the existence of a narrow cross-talk between pro-inflammatory and oxidative signals that can lead to neuronal damage and subsequent neurodegeneration. Further investigation about critical pathways crosslinking oxidative stress and inflammation will strength our knowlegde on this topic. Antioxid. Redox Signal. 28, 1626-1651.


Assuntos
Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Inflamação/metabolismo , Inflamação/patologia , Transdução de Sinais , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Oxirredução
15.
Mol Neurobiol ; 55(6): 5362-5376, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28936789

RESUMO

It has been shown that synergistic toxic effects of quinolinic acid (QUIN) and glutaric acid (GA), both in isolated nerve endings and in vivo conditions, suggest the contribution of these metabolites to neurodegeneration. However, this synergism still requires a detailed characterization of the mechanisms involved in cell damage during its occurrence. In this study, the effects of subtoxic concentrations of QUIN and/or GA were tested in neuronal cultures, co-cultures (neuronal cells + astrocytes), and mixed cultures (neuronal cells + astrocytes + microglia) from rat cortex and striatum. The exposure of different cortical and striatal cell cultures to QUIN + GA resulted in cell death and stimulated different markers of oxidative stress, including reactive oxygen species (ROS) formation; changes in the activity of antioxidant enzymes such as superoxide dismutase, catalase, and glutathione peroxidase; and depletion of endogenous antioxidants such as -SH groups and glutathione. The co-incubation of neuronal cultures with QUIN + GA plus the N-methyl-D-aspartate antagonist MK-801 prevented cell death but not ROS formation, whereas the antioxidant melatonin reduced both parameters. Our results demonstrated that QUIN and GA can create synergistic scenarios, inducing toxic effects on some parameters of cell viability via the stimulation of oxidative damage. Therefore, it is likely that oxidative stress may play a major causative role in the synergistic actions exerted by QUIN + GA in a variety of cell culture conditions involving the interaction of different neural types.


Assuntos
Glutaratos/toxicidade , Modelos Biológicos , Neurônios/metabolismo , Estresse Oxidativo , Ácido Quinolínico/toxicidade , Animais , Antioxidantes/metabolismo , Catalase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/patologia , Técnicas de Cocultura , Maleato de Dizocilpina/farmacologia , Feminino , Gliose/metabolismo , Gliose/patologia , Glutaratos/administração & dosagem , Glutationa/metabolismo , Melatonina/farmacologia , Neostriado/patologia , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Ácido Quinolínico/administração & dosagem , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo
18.
Neurotox Res ; 33(3): 634-640, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29313218

RESUMO

The mechanisms by which the heavy metal thallium (Tl+) produces toxicity in the brain remain unclear. Herein, isolated synaptosomal/mitochondrial P2 crude fractions from adult rat brains were exposed to Tl+ (5-250 µM) for 30 min. Three toxic endpoints were evaluated: mitochondrial dysfunction, lipid peroxidation, and Na+/K+-ATPase activity inhibition. Concentration-response curves for two of these endpoints revealed the optimum concentration of Tl+ to induce damage in this preparation, 5 µM. Toxic markers were also estimated in preconditioned synaptosomes incubated in the presence of the N-methyl-D-aspartate receptor antagonist kynurenic acid (KYNA, 50 µM), the cannabinoid receptor agonist WIN 55,212-2 (1 µM), or the antioxidant S-allyl-L-cysteine (SAC, 100 µM). All these agents prevented Tl+ toxicity, though SAC did it with lower efficacy. Our results suggest that energy depletion, oxidative damage, and Na+/K+-ATPase activity inhibition account for the toxic pattern elicited by Tl+ in nerve terminals. In addition, the efficacy of the drugs employed against Tl+ toxicity supports an active role of excitatory/cannabinoid and oxidative components in the toxic pattern elicited by the metal.


Assuntos
Encéfalo/ultraestrutura , Mitocôndrias/efeitos dos fármacos , Sinaptossomos/efeitos dos fármacos , Tálio/toxicidade , Animais , Benzoxazinas/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Cisteína/análogos & derivados , Cisteína/farmacologia , Relação Dose-Resposta a Droga , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Cinurênico/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Morfolinas/farmacologia , Naftalenos/farmacologia , Ratos , Ratos Wistar , ATPase Trocadora de Sódio-Potássio/metabolismo
19.
Neurotox Res ; 33(4): 837-845, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29124680

RESUMO

Levetiracetam (LVT) is a relatively novel antiepileptic drug (AED) known to act through binding with the synaptic vesicular 2A (SV2A) protein, thus modulating the presynaptic neurotransmitter release. The tryptophan metabolite quinolinic acid (QUIN) acts as an excitotoxin when its brain concentrations reach toxic levels under pathological conditions. Since increased neuronal excitability induced by QUIN recruits degenerative events in the brain, and novel AED is also expected to exert neuroprotective effects in their pharmacological profiles, in this work the effect of LVT (54 mg/kg, i.p., administered for seven consecutive days) was tested as a pretreatment against the toxicity evoked by the bilateral intrastriatal injection of QUIN (60 nmol/µl) to adult rats. QUIN increased the striatal levels of peroxidized lipids and carbonylated proteins as indexes of oxidative damage 24 h after its infusion. In addition, in synaptosomal fractions isolated from QUIN-lesioned rats 24 h after the toxin infusion, γ-aminobutyric acid (GABA) release was decreased, whereas glutamate (Glu) release was increased. QUIN also decreased motor activity and augmented the rate of cell damage at 7 days post-lesion. All these alterations were significantly prevented by pretreatment of rats with LVT. The results of this study show a neuroprotective role and antioxidant action of LVT against the brain damage induced by excitotoxic events.


Assuntos
Anticonvulsivantes/farmacologia , Corpo Estriado/efeitos dos fármacos , Levetiracetam/farmacologia , Neostriado/efeitos dos fármacos , Animais , Lesões Encefálicas/tratamento farmacológico , Masculino , Fármacos Neuroprotetores , Ácido Quinolínico/toxicidade , Ratos Wistar , Transmissão Sináptica/efeitos dos fármacos
20.
Neurotox Res ; 31(4): 532-544, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28092019

RESUMO

Several physiological events in the brain are regulated by the endocannabinoid system (ECS). While synthetic cannabinoid receptor (CBr) agonists such as WIN55,212-2 act directly on CBr, agents like URB597, a fatty acid amide hydrolase (FAAH) inhibitor, induce a more "physiological" activation of CBr by increasing the endogenous levels of the endocannabinoid anandamide (AEA). Herein, we compared the pre- and post-treatment efficacy of URB597 and WIN55,212-2 on different endpoints evaluated in the toxic model produced by the mitochondrial toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in mice. MPTP (40 mg/kg, s.c., single injection) decreased locomotor activity, depleted the striatal and nigral levels of dopamine (DA), augmented the levels of lipid peroxidation and protein carbonylation in both regions, decreased the striatal protein levels of tyrosine hydroxylase, and increased the striatal protein content of the subunit 1 (NR1) of the N-methyl-D-aspartate receptor (NMDAr). Both URB597 (0.3 mg/kg, i.p., once a day) and WIN55,212-2 (10 µg/kg, i.p., twice a day), administered for five consecutive days, either before or after the MPTP injection, prevented the alterations elicited by MPTP and downregulated NMDAr. Our results support a modulatory role of the ECS on the toxic profile exerted by MPTP in mice via the stimulation of antioxidant activity and the induction of NMDAr downregulation and hypofunction, and favor the stimulation of CBr as an effective experimental therapeutic strategy.


Assuntos
Benzamidas/farmacologia , Benzoxazinas/farmacologia , Carbamatos/farmacologia , Dopamina/metabolismo , Locomoção/efeitos dos fármacos , Intoxicação por MPTP/metabolismo , Morfolinas/farmacologia , Naftalenos/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Corpo Estriado/metabolismo , Regulação para Baixo/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Intoxicação por MPTP/prevenção & controle , Masculino , Camundongos , Oxirredução/efeitos dos fármacos , Carbonilação Proteica/efeitos dos fármacos , Substância Negra/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA