Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(12)2021 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-34208374

RESUMO

Previous studies have demonstrated that pioglitazone, a peroxisome proliferator-activated receptor gamma (PPARγ) agonist, inhibits ischemia-induced brain injury. The present study was conducted to examine whether pioglitazone can reduce impairment of behavioral deficits mediated by inflammatory-induced brain white matter injury in neonatal rats. Intraperitoneal (i.p.) injection of lipopolysaccharide (LPS, 2 mg/kg) was administered to Sprague-Dawley rat pups on postnatal day 5 (P5), and i.p. administration of pioglitazone (20 mg/kg) or vehicle was performed 5 min after LPS injection. Sensorimotor behavioral tests were performed 24 h after LPS exposure, and changes in biochemistry of the brain was examined after these tests. The results show that systemic LPS exposure resulted in impaired sensorimotor behavioral performance, reduction of oligodendrocytes and mitochondrial activity, and increases in lipid peroxidation and brain inflammation, as indicated by the increment of interleukin-1ß (IL-1ß) levels and number of activated microglia in the neonatal rat brain. Pioglitazone treatment significantly improved LPS-induced neurobehavioral and physiological disturbances including the loss of body weight, hypothermia, righting reflex, wire-hanging maneuver, negative geotaxis, and hind-limb suspension in neonatal rats. The neuroprotective effect of pioglitazone against the loss of oligodendrocytes and mitochondrial activity was associated with attenuation of LPS-induced increment of thiobarbituric acid reactive substances (TBARS) content, IL-1ß levels and number of activated microglia in neonatal rats. Our results show that pioglitazone prevents neurobehavioral disturbances induced by systemic LPS exposure in neonatal rats, and its neuroprotective effects are associated with its impact on microglial activation, IL-1ß induction, lipid peroxidation, oligodendrocyte production and mitochondrial activity.


Assuntos
Comportamento Animal , Encefalite/tratamento farmacológico , Mitocôndrias/patologia , Pioglitazona/uso terapêutico , Substância Branca/patologia , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Citocinas/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Encefalite/patologia , Feminino , Hipotermia Induzida , Lipopolissacarídeos , Microglia/efeitos dos fármacos , Microglia/patologia , Mitocôndrias/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/patologia , Pioglitazona/farmacologia , Gravidez , Ratos Sprague-Dawley , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Redução de Peso/efeitos dos fármacos , Substância Branca/efeitos dos fármacos
2.
J Toxicol Sci ; 39(1): 1-6, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24418703

RESUMO

1-phenyl-2-(1-pyrrolidinyl)-1-pentanone (α-PVP) is a new designer drug of the cathinone type. People who have taken drugs containing α-PVP or other synthetic cathinone reportedly lose consciousness, develop difficulty breathing, and at worst case, die. However, the mechanism underlying α-PVP-induced neurotoxicity is unknown. The objective of the present study was to investigate the effect of α-PVP on the central nervous system (CNS) and compare its neurotoxicity with that of methamphetamine (METH) in mice. Balb/c male mice (8 weeks old) were orally administered α-PVP (25 mg/kg) or METH (5 mg/kg). α-PVP induced a significant increase in locomotor activity, which occurred earlier than locomotor activity induced by METH. This increase was inhibited by the D1 receptor antagonist SCH23990 (50 µg/kg, i.p.) and the D2 receptor antagonist sulpiride (50 mg/kg, i.m.). The extracellular concentration of dopamine (DA) in the striatum, determined by in vivo microdialysis increased immediately after α-PVP administration. These results suggest that α-PVP stimulates DA release, causing an increase in locomotor activity, and that this stimulatory effect of α-PVP on CNS is mediated, at least in part, by the D1 and D2 receptors.


Assuntos
Estimulantes do Sistema Nervoso Central , Drogas Desenhadas/toxicidade , Atividade Motora/efeitos dos fármacos , Pentanonas/toxicidade , Pirrolidinas/toxicidade , Animais , Benzazepinas/farmacologia , Corpo Estriado/metabolismo , Dopamina/metabolismo , Antagonistas dos Receptores de Dopamina D2 , Neurônios Dopaminérgicos/fisiologia , Masculino , Metanfetamina/toxicidade , Camundongos , Pentanonas/farmacologia , Pirrolidinas/farmacologia , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D1/fisiologia , Receptores de Dopamina D2/fisiologia , Sulpirida/farmacologia
3.
J Toxicol Sci ; 39(1): 33-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24418707

RESUMO

The psychoactive recreational drug 3,4-methylenedioxymethamphetamine (MDMA) is widely abused. The fact that MDMA induces neurotoxic damage in serotonergic nerve endings is well known. However, the effects of MDMA on pregnant and neonatal animals remain unknown. Therefore, we studied the effects of gestational exposure to MDMA on birth, growth, and behavior of pups. Female BALB/c mice were orally administered either water (10 ml/kg) or MDMA (20 mg/10 ml/kg) from gestational day 1 to postnatal day (P) 21. MDMA did not affect the birth rate, but the survival rate of the pups significantly decreased. A significant reduction in body weight gain was observed in pups from MDMA-administered dams during P3-P21. Maternal MDMA treatment caused an attenuated cliff avoidance reaction and decreased motor function in the pups, as determined by the wire hanging test. These results suggest that MDMA treatment during pregnancy and lactation causes growth retardation and dysfunction of motor neurons in mouse pups.


Assuntos
Peso Corporal/efeitos dos fármacos , Crescimento e Desenvolvimento/efeitos dos fármacos , Drogas Ilícitas , Exposição Materna/efeitos adversos , Troca Materno-Fetal , N-Metil-3,4-Metilenodioxianfetamina/administração & dosagem , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Efeitos Tardios da Exposição Pré-Natal , Animais , Comportamento Animal/efeitos dos fármacos , Feminino , Idade Gestacional , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Neurônios Motores/efeitos dos fármacos , N-Metil-3,4-Metilenodioxianfetamina/metabolismo , Gravidez , Desempenho Psicomotor/efeitos dos fármacos , Tempo de Reação , Neurônios Serotoninérgicos/efeitos dos fármacos
4.
J Neuroinflammation ; 10: 45, 2013 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-23561827

RESUMO

BACKGROUND: Cyclooxygenase-2 (COX-2) is induced in inflammatory cells in response to cytokines and pro-inflammatory molecules, suggesting that COX-2 has a role in the inflammatory process. The objective of the current study was to examine whether celecoxib, a selective COX-2 inhibitor, could ameliorate lipopolysaccharide (LPS)-induced brain inflammation, dopaminergic neuronal dysfunction and sensorimotor behavioral impairments. METHODS: Intraperitoneal (i.p.) injection of LPS (2 mg/kg) was performed in rat pups on postnatal Day 5 (P5), and celecoxib (20 mg/kg) or vehicle was administered (i.p.) five minutes after LPS injection. Sensorimotor behavioral tests were carried out 24 h after LPS exposure, and brain injury was examined on P6. RESULTS: Our results showed that LPS exposure resulted in impairment in sensorimotor behavioral performance and injury to brain dopaminergic neurons, as indicated by loss of tyrosine hydroxylase (TH) immunoreactivity, as well as decreases in mitochondria activity in the rat brain. LPS exposure also led to increases in the expression of α-synuclein and dopamine transporter proteins and enhanced [3H]dopamine uptake. Treatment with celecoxib significantly reduced LPS-induced sensorimotor behavioral disturbances and dopaminergic neuronal dysfunction. Celecoxib administration significantly attenuated LPS-induced increases in the numbers of activated microglia and astrocytes and in the concentration of IL-1ß in the neonatal rat brain. The protective effect of celecoxib was also associated with an attenuation of LPS-induced COX-2+ cells, which were double labeled with TH + (dopaminergic neuron) or glial fibrillary acidic protein (GFAP) + (astrocyte) cells. CONCLUSION: Systemic LPS administration induced brain inflammatory responses in neonatal rats; these inflammatory responses included induction of COX-2 expression in TH neurons and astrocytes. Application of the COX-2 inhibitor celecoxib after LPS treatment attenuated the inflammatory response and improved LPS-induced impairment, both biochemically and behaviorally.


Assuntos
Inibidores de Ciclo-Oxigenase 2/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Atividade Motora/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Animais , Animais Recém-Nascidos , Western Blotting , Celecoxib , Complexo I de Transporte de Elétrons/fisiologia , Ensaio de Imunoadsorção Enzimática , Feminino , Imuno-Histoquímica , Inflamação/induzido quimicamente , Inflamação/psicologia , Interleucina-1beta/análise , Interleucina-1beta/metabolismo , Masculino , Mitocôndrias/metabolismo , Equilíbrio Postural/efeitos dos fármacos , Gravidez , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/metabolismo
5.
Toxicology ; 308: 96-103, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23567316

RESUMO

Brain inflammation in early life may enhance adult susceptibility to develop neurodegenerative disorders triggered by environmental toxins. Our previous studies show that perinatal lipopolysaccharide (LPS) exposure enhances adult susceptibility to rotenone-induced injury to the dopaminergic system in the substantia nigra (SN) of the adult rat brain. To further investigate the enhanced adult susceptibility by neonatal LPS exposure to rotenone neurotoxicity, we used our neonatal rat model of LPS exposure (1mg/kg, intracerebral injection in postnatal day 5, P5, neonatal rats) to examine the protein levels of α-synuclein and dopamine transporters (DAT) in the adult rat. By P70, rats from the saline- or LPS-exposed group were challenged with rotenone, a commonly used pesticide, through subcutaneous mini-pump infusion at a dose of 1.25mg/kg/day for 14 days. The accumulation of α-synuclein aggregation and increment of DAT protein content were found in the SN of LPS-exposed rats. Neonatal LPS exposure enhanced rotenone-stimulated accumulation of α-synuclein aggregation and increment in DAT protein expression in the cytoplasmic compartment of the SN, and in the synaptosomal compartment of the striatum of adult rats. Rotenone treatment also resulted in reduction of [(3)H]dopamine uptake and mitochondrial complex I activity in the striatum of rats with neonatal LPS exposure, but not in those without this exposure. The current study suggests possible roles of α-synuclein aggregate and DAT distribution in the cytoplasm and synaptosome triggered by environmental toxins in later life in the development of neurodegenerative disorders. Our model may be useful in studying mechanisms involved in the pathogenesis of nonfamilial Parkinson's disease and for developing potential therapeutic treatments for this disease.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Lipopolissacarídeos/toxicidade , Rotenona/toxicidade , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , alfa-Sinucleína/metabolismo , Animais , Animais Recém-Nascidos , Proteínas da Membrana Plasmática de Transporte de Dopamina/biossíntese , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley , Substância Negra/crescimento & desenvolvimento , alfa-Sinucleína/biossíntese
6.
Dev Neurosci ; 35(2-3): 155-71, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23446007

RESUMO

Brain inflammation via intracerebral injection with lipopolysaccharide (LPS) in early life has been shown to increase risks for the development of neurodegenerative disorders in adult rats. To determine if neonatal systemic LPS exposure has the same effects on enhancement of adult dopaminergic neuron susceptibility to rotenone neurotoxicity as centrally injected LPS does, LPS (2 µg/g body weight) was administered intraperitoneally into postnatal day 5 (P5) rats and when grown to P70, rats were challenged with rotenone, a commonly used pesticide, through subcutaneous minipump infusion at a dose of 1.25 mg/kg/day for 14 days. Systemically administered LPS can penetrate into the neonatal rat brain and cause acute and chronic brain inflammation, as evidenced by persistent increases in IL-1ß levels, cyclooxygenase-2 expression and microglial activation in the substantia nigra (SN) of P70 rats. Neonatal LPS exposure resulted in suppression of tyrosine hydroxylase (TH) expression, but not actual death of dopaminergic neurons in the SN, as indicated by the reduced number of TH+ cells and unchanged total number of neurons (NeuN+) in the SN. Neonatal LPS exposure also caused motor function deficits, which were spontaneously recoverable by P70. A small dose of rotenone at P70 induced loss of dopaminergic neurons, as indicated by reduced numbers of both TH+ and NeuN+ cells in the SN, and Parkinson's disease (PD)-like motor impairment in P98 rats that had experienced neonatal LPS exposure, but not in those without the LPS exposure. These results indicate that although neonatal systemic LPS exposure may not necessarily lead to death of dopaminergic neurons in the SN, such an exposure could cause persistent functional alterations in the dopaminergic system and indirectly predispose the nigrostriatal system in the adult brain to be damaged by environmental toxins at an ordinarily nontoxic or subtoxic dose and develop PD-like pathological features and motor dysfunction.


Assuntos
Encéfalo/patologia , Neurônios Dopaminérgicos/patologia , Inflamação/complicações , Lipopolissacarídeos/toxicidade , Rotenona/toxicidade , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Contagem de Células , Neurônios Dopaminérgicos/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Inflamação/induzido quimicamente , Inflamação/metabolismo , Masculino , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/fisiopatologia , Ratos , Ratos Sprague-Dawley , Desacopladores/toxicidade
7.
J Toxicol Sci ; 35(3): 375-81, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20519846

RESUMO

3,4-Methylenedioxymethamphetamine (MDMA) is widely abused as a psychoactive recreational drug. It is well known that MDMA induces neurotoxic damage of serotonergic nerve endings. Although drug abuse is increasing among youths, it is unclear whether recreational drugs affect the development of nerve growth. Thus, the present study examined the effect of recreational drugs, such as MDMA, 3,4-methylenedioxyamphetamine (MDA) and diphenylprolinol, a novel recreational drug with a similar chemical structure as that of psychoactive agent pipradrol, on nerve growth factor (NGF)-induced neurite outgrowth. These recreational drugs induced a dose-dependent cell death in PC12 cells. The IC(50) values of MDMA, MDA, R-diphenylprolinol and S-diphenylprolinol were 4.11 mM, 2.75 mM, 1.00 mM and 0.77 mM, respectively, at 24 hr. To examine the effects of these recreational drugs on NGF-induced neurite outgrowth, PC12 cells were treated with NGF together with MDMA, MDA, S-diphenylprolinol or R-diphenylprolinol at low toxic concentrations. The recreational drugs significantly suppressed neurite outgrowth of PC12 cells induced by NGF. The results suggest that these psychoactive recreational drugs may inhibit neurite growth and thus be implicated in their elicited neurotoxicity.


Assuntos
3,4-Metilenodioxianfetamina/toxicidade , Drogas Ilícitas/toxicidade , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Células PC12/efeitos dos fármacos , Prolina/análogos & derivados , Prolina/toxicidade , Animais , Depressão Química , Fator de Crescimento Neural/farmacologia , Células PC12/fisiologia , Ratos
8.
Brain Res ; 1108(1): 39-44, 2006 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-16828716

RESUMO

To investigate the role of heme oxygenase (HO) isozymes, we used siRNA technology to suppress HO-1 expression. HO-1 siRNA-transfected HT22 cells were vulnerable to hydrogen peroxide- and 4-hydroxynonenal-induced cytotoxicity. Biliverdin and bilirubin, degradative products of heme catalyzed by HO, protected HT22 cells from the insult of these oxidative stressors. These results suggest that inducible HO-1 plays a protective role against oxidative stress in HT22 cells.


Assuntos
Encéfalo/enzimologia , Heme Oxigenase-1/genética , Estresse Oxidativo/genética , Aldeídos/toxicidade , Animais , Encéfalo/fisiopatologia , Linhagem Celular Transformada , Regulação para Baixo/genética , Inativação Gênica/fisiologia , Peróxido de Hidrogênio/toxicidade , Camundongos , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/fisiopatologia , Fármacos Neuroprotetores/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Interferência de RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...