Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Ageing Res Rev ; 93: 102149, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38056504

RESUMO

Familial hypercholesterolemia (FH) is a metabolic condition caused mainly by a mutation in the low-density lipoprotein (LDL) receptor gene (LDLR), which is highly prevalent in the population. Besides being an important causative factor of cardiovascular diseases, FH has been considered an early risk factor for Alzheimer's disease. Cognitive and emotional behavioral impairments in LDL receptor knockout (LDLr-/-) mice are associated with neuroinflammation, blood-brain barrier dysfunction, impaired neurogenesis, brain oxidative stress, and mitochondrial dysfunction. Notably, today, LDLr-/- mice, a widely used animal model for studying cardiovascular diseases and atherosclerosis, are also considered an interesting tool for studying dementia. Here, we reviewed the main findings in LDLr-/- mice regarding the relationship between FH and brain dysfunctions and dementia development.


Assuntos
Doença de Alzheimer , Doenças Cardiovasculares , Hipercolesterolemia , Hiperlipoproteinemia Tipo II , Humanos , Animais , Camundongos , Hipercolesterolemia/epidemiologia , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Doenças Cardiovasculares/genética , Fatores de Risco , Hiperlipoproteinemia Tipo II/complicações , Hiperlipoproteinemia Tipo II/genética , Encéfalo/metabolismo , Cognição , Fatores de Risco de Doenças Cardíacas
2.
Biomed Pharmacother ; 142: 111196, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34210581

RESUMO

Oxidized LDL (oxLDL) plays a pivotal role on atherosclerosis development, mainly in the formation of lipid-laden macrophage "foam cells". As a consequence, substances that can modulate LDL oxidation have a pharmacological and therapeutic relevance. Based in previous findings showing the ability of Syzigium cumini leaf extract (ScExt) in preventing LDL oxidation in vitro, this study was aimed to assess the effects of ScExt on oxLDL-mediated toxicity in murine J774 macrophages-like cells. For biochemical analyses, LDL isolated from fresh human plasma and oxidized with CuSO4 was incubated with ScExt pre-treated macrophages. Our results demonstrated that ScExt was efficient in preventing the overproduction of reactive oxygen/nitrogen species (ROS/RNS), the loss of macrophage's viability and the foam cells formation induced by oxLDL. These protective effects of ScExt make it a promising antioxidant for future trials toward atherogenesis.


Assuntos
Antioxidantes/farmacologia , Aterosclerose/prevenção & controle , Macrófagos/efeitos dos fármacos , Extratos Vegetais/farmacologia , Folhas de Planta/química , Substâncias Protetoras/farmacologia , Syzygium/química , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Espumosas/citologia , Células Espumosas/efeitos dos fármacos , Humanos , Lipoproteínas LDL/toxicidade , Camundongos , Espécies Reativas de Oxigênio/metabolismo
3.
Colloids Surf B Biointerfaces ; 201: 111608, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33618084

RESUMO

Hypercholesterolemia has been linked to neurodegenerative disease development. Previously others and we demonstrated that high levels of plasma cholesterol-induced memory impairments and depressive-like behavior in mice. More recently, some evidence reported that a hypercholesterolemic diet led to motor alterations in rodents. Peripheral inflammation, blood-brain barrier (BBB) dysfunction, and neuroinflammation seem to be the connective factors between hypercholesterolemia and brain disorders. Herein, we aimed to investigate whether treatment with gold nanoparticles (GNPs) can prevent the inflammation, BBB disruption, and behavioral changes related to neurodegenerative diseases and depression, induced by hypercholesterolemic diet intake in mice. Adult Swiss mice were fed a standard or a high cholesterol diet for eight weeks and concomitantly treated with either vehicle or GNPs by the oral route. At the end of treatments, mice were subjected to behavioral tests. After that, the blood, liver, and brain structures were collected for biochemical analysis. The high cholesterol diet-induced an increase in the plasma cholesterol levels and body weight of mice, which were not modified by GNPs treatment. Hypercholesterolemia was associated with enhanced liver tumor necrosis factor- α (TNF-α), BBB dysfunction in the hippocampus and olfactory bulb, memory impairment, cataleptic posture, and depressive-like behavior. Notably, GNPs administration attenuated liver inflammation, BBB dysfunction, and improved behavioral and memory deficits in hypercholesterolemic mice. Also, GNPs increased mitochondrial complex I activity in the prefrontal cortex of mice. It is worth highlight that GNPs' administration did not cause toxic effects in the liver and kidney of mice. Overall, our results indicated that GNPs treatment potentially mitigated peripheral, brain, and memory impairments related to hypercholesterolemia.


Assuntos
Hipercolesterolemia , Nanopartículas Metálicas , Doenças Neurodegenerativas , Animais , Ouro , Hipercolesterolemia/tratamento farmacológico , Camundongos , Nanotecnologia
4.
Neurochem Res ; 46(1): 120-130, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32285377

RESUMO

Most pharmacological studies concerning the beneficial effects of organoselenium compounds have focused on their ability to mimic glutathione peroxidase (GPx). However, mechanisms other than GPx-like activity might be involved on their biological effects. This study was aimed to investigate and compare the protective effects of two well known [(PhSe)2 and PhSeZnCl] and two newly developed (MRK Picolyl and MRK Ester) organoselenium compounds against oxidative challenge in cultured neuronal HT22 cells. The thiol peroxidase and oxidase activities were performed using the glutathione reductase (GR)-coupled assay. In order to evaluate protective effects of the organoselenium compounds against oxidative challenge in neuronal HT22 cells, experiments based on glutamate-induced oxytosis and SIN-1-mediated peroxynitrite generation were performed. The thiol peroxidase activities of the studied organoselenium compounds were smaller than bovine erythrocytes GPx enzyme. Besides, (PhSe)2 and PhSeZnCl showed higher thiol peroxidase and lower thiol oxidase activities compared to the new compounds. MRK Picolyl and MRK Ester, which showed lower thiol peroxidase activity, showed higher thiol oxidase activity. Both pre- or co-treatment with (PhSe)2, PhSeZnCl, MRK Picolyl and MRK Ester protected HT22 cells against glutamate-induced cytotoxicity. (PhSe)2 and MRK Picolyl significantly prevented peroxinitrite-induced dihydrorhodamine oxidation, but this effect was observed only when HT22 were pre-treated with these compounds. The treatment with (PhSe)2 increased the protein expression of antioxidant defences (Prx3, CAT and GCLC) in HT22 cells. Taking together, our results suggest that the biological effects elicited by these compounds are not directly related to their GPx-mimetic and thiol oxidase activities, but might be linked to the up-regulation of endogenous antioxidant defences trough their thiol-modifier effects.


Assuntos
Antioxidantes/farmacologia , Neurônios/efeitos dos fármacos , Compostos Organosselênicos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Catalase/metabolismo , Bovinos , Linhagem Celular , Glutamato-Cisteína Ligase/metabolismo , Glutationa Peroxidase/metabolismo , Proteínas de Homeodomínio/metabolismo , Camundongos
5.
Mol Neurobiol ; 58(2): 735-749, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33011857

RESUMO

Methylglyoxal (MGO) is an endogenous toxin, mainly produced as a by-product of glycolysis that has been associated to aging, Alzheimer's disease, and inflammation. Cell culture studies reported that MGO could impair the glyoxalase, thioredoxin, and glutathione systems. Thus, we investigated the effect of in vivo MGO administration on these systems, but no major changes were observed in the glyoxalase, thioredoxin, and glutathione systems, as evaluated in the prefrontal cortex and the hippocampus of mice. A previous study from our group indicated that MGO administration produced learning/memory deficits and depression-like behavior. Confirming these findings, the tail suspension test indicated that MGO treatment for 7 days leads to depression-like behavior in three different mice strains. MGO treatment for 12 days induced working memory impairment, as evaluated in the Y maze spontaneous alternation test, which was paralleled by low dopamine and serotonin levels in the cerebral cortex. Increased DARPP32 Thr75/Thr34 phosphorylation ratio was observed, suggesting a suppression of phosphatase 1 inhibition, which may be involved in behavioral responses to MGO. Co-treatment with a dopamine/noradrenaline reuptake inhibitor (bupropion, 10 mg/kg, p.o.) reversed the depression-like behavior and working memory impairment and restored the serotonin and dopamine levels in the cerebral cortex. Overall, the cerebral cortex monoaminergic system appears to be a preferential target of MGO toxicity, a new potential therapeutic target that remains to be addressed.


Assuntos
Depressão/fisiopatologia , Inibidores da Captação de Dopamina/farmacologia , Dopamina/deficiência , Memória de Curto Prazo , Norepinefrina/metabolismo , Aldeído Pirúvico/efeitos adversos , Animais , Bupropiona/farmacologia , Dopamina/metabolismo , Feminino , Glutationa/metabolismo , Imobilização , Memória de Curto Prazo/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Aldeído Pirúvico/administração & dosagem , Serotonina/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
6.
Sci Rep ; 9(1): 20119, 2019 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-31882804

RESUMO

Zika virus (ZIKV) has a strong tropism for the nervous system and has been related to post-infection neurological syndromes. Once neuronal cells are infected, the virus is capable of modulating cell metabolism, leading to neurotoxicity and cellular death. The negative effect of ZIKV in neuron cells has been characterized. However, the description of molecules capable of reversing these cytotoxic effects is still under investigation. In this context, it has been largely demonstrated that docosahexaenoic acid (DHA), an omega-3 polyunsaturated fatty acid, is highly neuroprotective. Here, we hypothesized that DHA's neuroprotective proprieties could have an influence on ZIKV-induced neurotoxicity in SH-SY5Y cells. Our data showed that pre-treatment of SH-SY5Y cells with DHA increased the cell viability and proliferation in ZIKV-infected cells. Moreover, DHA triggered an anti-inflammatory response in those infected cells. Besides, DHA was capable of restoring mitochondria function and number in ZIKV-infected SH-SY5Y cells. In addition, cells pre-treated with DHA prior to ZIKV infection presented a lower viral load at different times of infection. Taking together, these results demonstrated that DHA has a potential anti-inflammatory and neuroprotective effect against ZIKV infection in these neuron-like cells and could be a useful tool in the treatment against this virus.


Assuntos
Anti-Inflamatórios/farmacologia , Ácidos Graxos Ômega-3/farmacologia , Fármacos Neuroprotetores/farmacologia , Zika virus/efeitos dos fármacos , Zika virus/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Efeito Citopatogênico Viral/efeitos dos fármacos , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Replicação Viral/efeitos dos fármacos , Infecção por Zika virus/virologia
7.
Redox Biol ; 20: 118-129, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30308475

RESUMO

Oxidative stress and mitochondrial dysfunction are critical events in neurodegenerative diseases; therefore, molecules that increase cellular antioxidant defenses represent a future pharmacologic strategy to counteract such conditions. The aim of this study was to investigate the potential protective effect of (PhSe)2 on mouse hippocampal cell line (HT22) exposed to tert-BuOOH (in vitro model of oxidative stress), as well as to elucidate potential mechanisms underlying this protection. Our results showed that tert-BuOOH caused time- and concentration-dependent cytotoxicity, which was preceded by increased oxidants production and mitochondrial dysfunction. (PhSe)2 pre-incubation significantly prevented these cytotoxic events and the observed protective effects were paralleled by the upregulation of the cellular glutathione-dependent antioxidant system: (PhSe)2 increased GSH levels (> 60%), GPx activity (6.9-fold) and the mRNA expression of antioxidant enzymes Gpx1 (3.9-fold) and Gclc (2.3-fold). Of note, the cytoprotective effect of (PhSe)2 was significantly decreased when cells were treated with mercaptosuccinic acid, an inhibitor of GPx, indicating the involvement of GPx modulation in the observed protective effect. In summary, the present findings bring out a new action mechanism concerning the antioxidant properties of (PhSe)2. The observed upregulation of the glutathione-dependent antioxidant system represents a future pharmacologic possibility that goes beyond the well-known thiol-peroxidase activity of this compound.


Assuntos
Derivados de Benzeno/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Compostos Organosselênicos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Animais , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Glutationa/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Camundongos , Modelos Biológicos , Oxidantes/biossíntese , Oxirredução/efeitos dos fármacos
8.
Behav Brain Res ; 359: 648-656, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30287273

RESUMO

While chronic high-fat feeding has long been associated with the rising incidence of obesity/type 2 diabetes, recent evidence has established that it is also associated with deficits in hippocampus-dependent memory. In this regard, environmental enrichment (EE) is an animal housing technique composed of increased space, physical activity, and social interactions, which in turn increases sensory, cognitive, motor, and social stimulation. EE leads to improved cerebral health as defined by increased neurogenesis, enhanced learning and memory and resistance to external cerebral insults. In the present study, the impacts of environmental enrichment (EE) on Swiss mice fed a high-fat, cholesterol-enriched diet (HFECD; 20% fat and 1.5% cholesterol) were investigated. Here, we demonstrated that EE, when initiated 4 weeks after the beginning of HFECD in mice, prevents HFECD-induced spatial memory and object recognition impairment, which were tested in T-maze and object recognition tests. Although EE did not affect HFECD-induced weight gain or hypercholesterolaemia, it improved glucose tolerance. On the other hand, EE was unable to mitigate a decrease in brain-derived neurotrophic factor (BDNF) and IL-6 hippocampal levels induced by the HFECD. Overall, while our results reinforce the positive and neuroprotective effects of EE on cognition they do not support a role for EE in preventing the neurochemical changes induced by the HFECD. Based on clinical observations that nondiabetic individuals with mild forms of impaired glucose tolerance have a higher risk of cognitive impairments, one can speculate about the connection between the effects of EE on glucose intolerance and its effects on cognition.


Assuntos
Colesterol/efeitos adversos , Disfunção Cognitiva/terapia , Dieta Hiperlipídica/efeitos adversos , Meio Ambiente , Abrigo para Animais , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Intolerância à Glucose/etiologia , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Intolerância à Glucose/terapia , Hipocampo/metabolismo , Hipocampo/patologia , Hipercolesterolemia/etiologia , Hipercolesterolemia/metabolismo , Hipercolesterolemia/patologia , Hipercolesterolemia/psicologia , Interleucina-6/metabolismo , Masculino , Camundongos , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia , Obesidade/psicologia , Distribuição Aleatória , Reconhecimento Psicológico , Memória Espacial
9.
Neurotox Res ; 33(3): 549-559, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29164519

RESUMO

Atorvastatin is a 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor with cholesterol-lowering, anti-inflammatory, and antioxidant properties. Increasing evidence show atorvastatin acts as a protective agent against insults in the central nervous system (CNS). The regular use of statins has been associated with a reduced risk of Parkinson's disease (PD) development. Here, we evaluated early events involved in the neurotoxicity induced by intranasal (i.n.) infusion of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in rats and the potential of atorvastatin to prevent these early toxic events. Male Wistar rats were pretreated orally with atorvastatin (10 mg/kg/day) or vehicle once a day during seven consecutive days. Twenty-four hours after atorvastatin administration, animals received a single bilateral i.n. infusion of MPTP (1 mg/nostril), and 6 h later, the striatum and the hippocampus were collected to evaluate early oxidative stress parameters and inflammatory cytokines. Atorvastatin prevented MPTP-induced increase in reactive species (RS) generation and in glutathione levels in the striatum. Atorvastatin also prevented the reduction in mitochondrial respiratory chain complex I and II activities evoked by MPTP in the striatum. Atorvastatin per se reduced the levels of the cytokines TNF-α and IL-1ß, and surprisingly, it reduced IL-10 and nerve growth factor levels in the striatum. However, the anti-inflammatory IL-10 levels increased in the striatum following atorvastatin plus MPTP treatment. These effects were not observed in the hippocampus. Our findings reinforce and extend the notion of the neuroprotective effects of atorvastatin in a PD model and indicate the modulation of oxidative and inflammatory responses as the mechanisms associated with therapeutic action of atorvastatin in PD.


Assuntos
Atorvastatina/administração & dosagem , Citocinas/metabolismo , Intoxicação por MPTP/prevenção & controle , Fármacos Neuroprotetores/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , Análise de Variância , Animais , Vias de Administração de Medicamentos , Esquema de Medicação , Complexo II de Transporte de Elétrons/metabolismo , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , Masculino , Neurotoxinas/administração & dosagem , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
10.
Neurochem Res ; 43(2): 387-396, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29134401

RESUMO

The contribution of oxidative stress to the pathophysiology of depression has been described in numerous studies. Particularly, an increased production of reactive oxygen species (ROS) caused by mitochondrial dysfunction can lead to neuronal cell death. Human neuroblastoma SH-SY5Y cells were used to investigate the neuroprotective effect of the antidepressant duloxetine against rotenone-induced oxidative stress. SH-SY5Y cells were pretreated with duloxetine (1-5 µM) for 24 h followed by a 24-h rotenone exposure (10 µM). The phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) inhibitor LY294002 (10 µM) and the heme oxygenase 1 (HO-1) inhibitor zinc protoporphyrin IX-ZnPP (5 µM) were added to cultures 1 h prior duloxetine treatments. After treatments cell viability and ROS generation were assessed. NF-E2-related factor-2 (Nrf2) nuclear translocation was assessed by immunofluorescent staining after 4 and 8 h of duloxetine incubation. Furthermore, the Nrf2 and HO-1 mRNA expression was carried out after 4-48 h of duloxetine treatment by qRT-PCR. Duloxetine pretreatment antagonized rotenone-induced overproduction of ROS and cell death in SH-SY5Y cells. In addition, a 1-h pretreatment with LY294002 abolished duloxetine's protective effect. Duloxetine also induced nuclear translocation of the Nrf2 and the expression of its target gene, HO-1. Finally, the HO-1 inhibitor, ZnPP, suppressed the duloxetine protective effect. Overall, these results indicate that the mechanism of duloxetine neuroprotective action against oxidative stress and cell death might rely on the Akt/Nrf2/HO-1 pathways.


Assuntos
Morte Celular/efeitos dos fármacos , Cloridrato de Duloxetina/farmacologia , Neuroblastoma/tratamento farmacológico , Espécies Reativas de Oxigênio/metabolismo , Rotenona/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Heme Oxigenase-1/metabolismo , Humanos , Fármacos Neuroprotetores/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
11.
Toxicol In Vitro ; 42: 273-280, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28461233

RESUMO

Thiol homeostasis has a critical role in the maintenance of proper cellular functions and survival, being coordinated by the action of several reductive enzymes, including glutathione (GSH)/glutathione reductase (GR) and thioredoxin (Trx)/thioredoxin reductase (TrxR) systems. Here, we investigated the effects of the GR inhibitor 2-acetylamino-3-[4-(2-acetylamino-2-carboxyethylsulfanylthiocarbonylamino)phenylthiocarbamoylsulfanyl]propionic acid (2-AAPA) on the activity of thiol reductases (GR and TrxR), redox balance and mitochondrial function of A172 glioblastoma cells. 2-AAPA inhibited cell GR (IC50=6.7µM) and TrxR (IC50=8.7µM). A significant decrease in the cellular ability to decompose cumene hydroperoxide was observed and associated to a greater susceptibility to this peroxide. The redox state of peroxiredoxins (Prx1, Prx2 and Prx3) was markedly shifted to dimer 30min after treatment with 100µM 2-AAPA, an event preceding 2-AAPA-induced decrease in cell viability. Furthermore, mitochondrial function was also severely impaired, leading to a decrease in the respiratory control ratio, reserve capacity, and ATP synthesis-coupled respiration, as well as an increase in mitochondrial membrane potential. Our results indicate that inhibition of GR and TrxR activities, disruption of the ability to detoxify peroxides, increased oxidation of Prxs, as well as compromised mitochondrial function represent early events mediating 2-AAPA toxicity to A172 glioblastoma cells.


Assuntos
Acetilcisteína/análogos & derivados , Antineoplásicos/farmacologia , Glutationa Redutase/antagonistas & inibidores , Tiocarbamatos/farmacologia , Tiorredoxina Dissulfeto Redutase/antagonistas & inibidores , Acetilcisteína/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glutationa Redutase/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Peroxirredoxinas/metabolismo , Tiorredoxina Dissulfeto Redutase/metabolismo
12.
Toxicol In Vitro ; 38: 19-26, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27989546

RESUMO

Emerging evidence has pointed to mercury exposure as a risk factor for hypertension, atherosclerosis, myocardial infarction and coronary heart disease. However, the underlying mechanisms are not well understood. This study investigated potential toxic effects of low concentrations of methylmercury (MeHg) in cultured bovine aortic endothelial cells (BAECs) and the possible involvement of reactive species, particularly superoxide anion, in mediating such toxicity. MeHg treatment increased the oxidation of 2',7'-dichlorodihydrofluorescein diacetate (a general probe for reactive species) and dihydroethidium, a specific probe for superoxide anion. MeHg-induced 2',7'-dichlorodihydrofluorescein diacetate and dihydroethidium oxidations were significantly decreased by apocynin, an inhibitor of the enzyme NADPH oxidase, which represents a main source of superoxide anion in endothelial cells. MeHg treatment significantly disrupted mitochondrial membrane potential and this event was also reversed by apocynin. MeHg treatment also decreased glutathione levels and this event preceded glutathione peroxidase inhibition, which was observed only at 24h after treatment. These results indicate that MeHg induces oxidative stress in cultured BAECs and that this event is related to the production of superoxide anion. Moreover, the observed protective effects of apocynin in BAECs suggest the potential involvement of NADPH-oxidase in MeHg-induced endothelial dysfunction, which represents a pivotal event in most cardiovascular diseases.


Assuntos
Células Endoteliais/efeitos dos fármacos , Compostos de Metilmercúrio/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Superóxidos/metabolismo , Acetofenonas/farmacologia , Animais , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/metabolismo , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , NADPH Oxidases/antagonistas & inibidores
13.
Behav Brain Res ; 311: 31-38, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27185735

RESUMO

Although epidemiological studies have reported an association between hypercholesterolemia and mood disorders, there is a lack of data regarding depressive-like behavior in animal models of hypercholesterolemia. To address these questions, we assessed depressive-like behavior and hippocampal and cortical monoaminergic metabolism in three-month-old, low-density lipoprotein receptor knockout (LDLr(-/-)) and C57BL/6 wild-type mice. The LDLr(-/-) mice exhibited depressive-like behavior in the sucrose preference test, splash test, and tail suspension test. Increased monoamine oxidase (MAO) A and B activity was evidenced in the hippocampus of LDLr(-/-) mice. Furthermore, to address whether or not cholesterol modulates MAO activity, we exposed SH-SY5Y human neuroblastoma cells to human isolated low-density lipoprotein (LDL). Notably, LDL increased the activity of MAO-A and stimulated the reactive species generation in vitro. These findings indicate that depressive-like behavior in hypercholesterolemic mice is accompanied by alterations in the monoaminergic metabolism, providing new evidence about the association between hypercholesterolemia and depression.


Assuntos
Depressão/metabolismo , Hipercolesterolemia/metabolismo , Hipercolesterolemia/psicologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monoaminoxidase/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética
14.
Mol Neurobiol ; 53(2): 1280-1295, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25619973

RESUMO

Succinobucol (succinyl ester of probucol) is a lipid-lowering compound with anti-inflammatory and antioxidant properties. Recent experimental evidence has highlighted the potential neuroprotective effects of succinobucol. In the present study, cultured neuroblastoma (SH-SY5Y) cells were used to investigate mechanisms mediating the potential protective effect of succinobucol against mitochondrial metabolic impairment and oxidative stress induced by 3-nitropropionic acid (3-NP), a succinate dehydrogenase inhibitor that has been used in experimental models of the Huntington disease (HD). 3-NP decreased cellular viability after 24 h of incubation. This decline in cellular viability was preceded by (i) reduced mitochondrial complex II activity, (ii) increased reactive species generation, (iii) decreased mitochondrial membrane potential (ΔΨm), and (iv) diminished glutathione (GSH) levels. Succinobucol pretreatment (6 days) significantly prevented 3-NP-induced loss of cellular viability, generation of reactive oxygen species, and decrease of ΔΨm. However, succinobucol pretreatment did not protect against 3-NP-induced inhibition of mitochondrial complex II activity, pointing to the mitigation of secondary events resultant from mitochondrial complex II inhibition. Succinobucol pretreatment (6 days) significantly increased (50 %) the levels of GSH in SH-SY5Y cells, and this event was paralleled by significant increases in glutamate cysteine ligase messenger RNA (mRNA) expression and activity (GCL; the first enzyme in the GSH biosynthesis). The present findings are the first to show that succinobucol increases GSH levels via upregulation of GCL activity (possibly through the activation of the nuclear (erythroid-derived 2)-related factor (Nrf2)/antioxidant response element (ARE) pathway), displaying protective effects against mitochondrial dysfunction-derived oxidative stress.


Assuntos
Glutamato-Cisteína Ligase/metabolismo , Glutationa/metabolismo , Hipolipemiantes/farmacologia , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Probucol/análogos & derivados , Regulação para Cima/efeitos dos fármacos , Butionina Sulfoximina/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Glutamato-Cisteína Ligase/genética , Glutationa Peroxidase/metabolismo , Humanos , Hidroquinonas/farmacologia , Mitocôndrias/efeitos dos fármacos , Nitrocompostos , Probucol/farmacologia , Propionatos , Substâncias Protetoras/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Tempo
16.
Biochimie ; 105: 172-81, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25038571

RESUMO

Elevated levels of oxidized low density lipoprotein (oxLDL) are considered to be one of the major risk factors for atherosclerosis and cardiovascular morbidity. The early stages of atherosclerosis are initiated by the accumulation of oxLDL and the induction of toxic effects on endothelial cells, resulting in endothelial dysfunction. The aim of this study was to investigate how diphenyl diselenide (DD), an organoselenium compound, protect vascular endothelial cells against the toxic effects of oxLDL in vitro. Our data showed that the treatment of bovine endothelial aortic cells (BAEC) with DD (0.1-1 µM) for 24 h protected from oxLDL-induced reactive species (RS) production and reduced glutathione (GSH) depletion. Moreover, DD (1 µM) per se improved the maximal mitochondrial respiratory capacity and prevented oxLDL-induced mitochondrial damage. In addition, DD could prevent apoptosis induced by oxLDL in BAEC. Results from this study may provide insight into a possible molecular mechanism underlying DD suppression of oxLDL-mediated vascular endothelial dysfunction.


Assuntos
Aterosclerose/tratamento farmacológico , Derivados de Benzeno/administração & dosagem , Células Endoteliais/efeitos dos fármacos , Compostos Organosselênicos/administração & dosagem , Substâncias Protetoras/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Aterosclerose/etiologia , Aterosclerose/metabolismo , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais/patologia , Glutationa/metabolismo , Humanos , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/toxicidade , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/metabolismo , Espécies Reativas de Oxigênio/metabolismo
17.
J Alzheimers Dis ; 41(1): 43-60, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24577472

RESUMO

Familial hypercholesterolemia is caused by inherited genetic abnormalities that directly or indirectly affect the function of the low-density lipoprotein (LDL) receptor. This condition is characterized by defective catabolism of LDL which results in increased plasma cholesterol concentrations and premature coronary artery disease. Nevertheless, there is increasing preclinical and clinical evidence indicating that familial hypercholesterolemia subjects show a particularly high incidence of mild cognitive impairment. Moreover, the LDL receptor (LDLr) has been implicated as the main central nervous system apolipoprotein E receptor that regulates amyloid deposition in distinct mouse models of ß-amyloidosis. In this regard, herein we hypothesized that the lack of LDLr would enhance the susceptibility to amyloid-ß-(Aß)-induced neurotoxicity in mice. Using the acute intracerebroventricular injection of aggregated Aß(1-40) peptide (400 pmol/mouse), a useful approach for the investigation of molecular mechanisms involved in Aß toxicity, we observed oxidative stress, neuroinflammation, and neuronal membrane damage within the hippocampus of C57BL/6 wild-type mice, which were associated with spatial reference memory and working memory impairments. In addition, our data show that LDLr knockout (LDLr(-/-)) mice, regardless of Aß treatment, displayed memory deficits and increased blood-brain barrier permeability. Nonetheless, LDLr(-/-) mice treated with Aß(1-40) peptide presented increased acetylcholinesterase activity, astrogliosis, oxidative imbalance, and cell permeability within the hippocampus in comparison with Aß(1-40)-treated C57BL/6 wild-type mice. Overall, the present study shows that the lack of LDLr increases the susceptibility to Aß-induced neurotoxicity in mice providing new evidence about the crosslink between familial hypercholesterolemia and cognitive impairment.


Assuntos
Amiloidose/fisiopatologia , Hipocampo/fisiopatologia , Transtornos da Memória/fisiopatologia , Córtex Pré-Frontal/fisiopatologia , Receptores de LDL/metabolismo , Acetilcolinesterase/metabolismo , Peptídeos beta-Amiloides , Amiloidose/complicações , Amiloidose/patologia , Animais , Antioxidantes/metabolismo , Astrócitos/patologia , Astrócitos/fisiologia , Barreira Hematoencefálica/fisiopatologia , Permeabilidade Capilar/fisiologia , Permeabilidade da Membrana Celular/fisiologia , Modelos Animais de Doenças , Gliose/patologia , Gliose/fisiopatologia , Hipocampo/patologia , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Memória de Curto Prazo/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroimunomodulação/fisiologia , Estresse Oxidativo/fisiologia , Fragmentos de Peptídeos , Córtex Pré-Frontal/patologia , Receptores de LDL/genética , Memória Espacial/fisiologia , Superóxido Dismutase/metabolismo
18.
J Neural Transm (Vienna) ; 121(4): 415-26, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24166183

RESUMO

Epidemiological studies have indicated hypercholesterolemia in midlife as a risk factor for dementia in later life, bringing cholesterol to the forefront of Alzheimer's disease research. Herein, we modeled mild hypercholesterolemia in mice to evaluate biochemical and behavioral alterations linked to hypercholesterolemia. Swiss mice were fed a high fat/cholesterol diet (20 % fat and 1.25 % cholesterol) for an 8-week period (from 12 to 18 weeks old) and were tested on the object location, forced swimming and elevated plus-maze tasks. We also investigated hypercholesterolemia-induced changes on acetylcholinesterase (AChE) activity, oxidative damage, amyloid precursor protein (APP) processing and blood brain barrier (BBB) integrity within the prefrontal cortex and hippocampus. It was found that increased AChE activity within the prefrontal cortex and hippocampus is an early event associated with hypercholesterolemia-induced short-term memory impairments. We observed no signs of antioxidant imbalance and/or oxidative damage or changes in cortical and hippocampal densities of beta-site amyloid precursor protein-cleaving enzyme 1 and aquaporin-4, biomarkers of APP processing and BBB integrity, respectively. In addition, we treated SH-SY5Y human neuroblastoma cells with low-density lipoprotein (LDL) cholesterol in an attempt to manipulate cell cholesterol content. Notably, LDL cholesterol increased in a dose-dependent manner the activity of AChE in SH-SY5Y cells. The present findings provide new evidence that increased AChE activity within the prefrontal cortex and hippocampus is an early event associated with hypercholesterolemia-induced cognitive impairments.


Assuntos
Acetilcolinesterase/metabolismo , Hipercolesterolemia/complicações , Transtornos da Memória/etiologia , Transtornos da Memória/metabolismo , Regulação para Cima/fisiologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Análise de Variância , Animais , Aquaporina 4/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Glicemia/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Glutationa/metabolismo , Glutationa Redutase/metabolismo , Hipocampo/enzimologia , Lipídeos/sangue , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Neuroblastoma/patologia , Córtex Pré-Frontal/enzimologia , Natação/psicologia , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
19.
J Pharm Pharmacol ; 66(3): 387-97, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24219623

RESUMO

OBJECTIVES: The present work aimed to investigate the effect of (PhSe)2 on cardiovascular age-related oxidative stress in hypercholesterolemic mice. METHODS: To this end, LDL receptor knockout (LDLr(-/-) ) mice, 3 months (young adult) and 12 months (middle-aged) old, were orally treated with (PhSe)2 . KEY FINDINGS: Hypercholesterolemia, regardless of age, impaired the mitochondrial antioxidant defence in the cardiac tissue, which was characterized by a decline in mitochondrial aortic glutathione (GSH) levels and increased reactive oxygen species production in the heart. (PhSe)2 treatment improved GSH levels, thioredoxin reductase (TRxR) and GSH reductase (GR) activity, and decreased malondialdehyde levels in the heart of young adult LDLr(-/-) mice. Moreover, (PhSe)2 increased GPx activity in both age groups, and GR activity in the aorta of middle-aged LDLr(-/-) mice. CONCLUSIONS: Therefore, (PhSe)2 enhances the antioxidant defences in the cardiovascular system of LDLr(-/-) mice, which could explain its success as an anti-atherogenic compound.


Assuntos
Antioxidantes/farmacologia , Derivados de Benzeno/farmacologia , Coração/efeitos dos fármacos , Hipercolesterolemia/metabolismo , Miocárdio/metabolismo , Compostos Organosselênicos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Receptores de LDL/metabolismo , Fatores Etários , Animais , Antioxidantes/metabolismo , Aorta , Aterosclerose/prevenção & controle , Glutationa/metabolismo , Glutationa Redutase/metabolismo , Malondialdeído/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico
20.
Chem Biol Interact ; 206(1): 18-26, 2013 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23954720

RESUMO

Interest in organoselenide chemistry and biochemistry has increased in the past three decades, mainly due to their chemical and biological activities. Here, we investigated the protective effect of the organic selenium compound diphenyl diselenide (PhSe)2 (5 µmol/kg), in a mouse model of methylmercury (MeHg)-induced brain toxicity. Our group has previously demonstrated that the oral and repeated administration (21 days) of MeHg (40 mg/L) induced MeHg brain accumulation at toxic concentrations, and a pattern of severe cortical and cerebellar biochemical and behavioral. In order to assess neurotoxicity, the neurochemical parameters, namely, mitochondrial complexes I, II, II-III and IV, glutathione peroxidase (GPx) and glutathione reductase (GR) activities, the content of thiobarbituric acid-reactive substances (TBA-RS), 8-hydroxy-2'-deoxyguanosine (8-OHdG), and brain-derived neurotrophic factor (BDNF), as well as, metal deposition were investigated in mouse cerebral cortex. Cortical neurotoxicity induced by brain MeHg deposition was characterized by the reduction of complexes I, II, and IV activities, reduction of GPx and increased GR activities, increased TBA-RS and 8-OHdG content, and reduced BDNF levels. The daily treatment with (PhSe)2 was able to counteract the inhibitory effect of MeHg on mitochondrial activities, the increased oxidative stress parameters, TBA-RS and 8-OHdG levels, and the reduction of BDNF content. The observed protective (PhSe)2 effect could be linked to its antioxidant properties and/or its ability to reduce MeHg deposition in brain, which was here histochemically corroborated. Altogether, these data indicate that (PhSe)2 could be consider as a neuroprotectant compound to be tested under neurotoxicity.


Assuntos
Antineoplásicos/farmacologia , Derivados de Benzeno/farmacologia , Córtex Cerebral/efeitos dos fármacos , Modelos Animais de Doenças , Fármacos Neuroprotetores/farmacologia , Compostos Organosselênicos/farmacologia , Animais , Antineoplásicos/química , Derivados de Benzeno/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Masculino , Compostos de Metilmercúrio/química , Compostos de Metilmercúrio/farmacologia , Camundongos , Fármacos Neuroprotetores/química , Compostos Organosselênicos/química , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA