Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Appl Toxicol ; 43(11): 1594-1603, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37189300

RESUMO

The metabolism of polycyclic aromatic hydrocarbons (PAHs) and the elimination kinetics of their mono-hydroxylated metabolites (OH-PAHs) following single exposure to different combinations of four PAHs (PAH4) were studied. Male Sprague-Dawley rats were orally exposed to a single dose of benzo[a]pyrene (B[a]P) or PAH2 (B[a]P + chrysene), PAH3 (B[a]P + chrysene + benz[a]anthracene), and PAH4 (B[a]P + chrysene + B[a]A + benzo[b]fluoranthene) with each combination adjusted to the same dose of individual compound. OH-PAHs including 3-hydroxybenzo[a]pyrene, 3-hydroxychrysene, 3-hydroxybenz[a]anthracene, and 1-hydroxypyrene (1-OHP) were detected in serum and urine samples collected at six intervals over a 72-h period post-dosing. The hepatic mRNA levels of cytochrome P450 (CYPs) were determined to ascertain the expression induction of PAHs metabolic enzymes. Results showed OH-PAHs (except 1-OHP) peaked within 8 h in serum and were excreted from urine within 24-48 h. The serum and urinary concentration of 3-hydroxybenzo[a]pyrene was significantly increased after PAH4 exposure compared with other PAHs combinations. Inversely, urinary concentration of 3-hydroxychrysene was decreased after PAH4 exposure, and the kinetics of 3-hydroxybenz[a]anthracene or 1-OHP were not different depending on the PAHs combinations. Also, CYPs were markedly induced by PAHs. Notably, the induction levels of CYP1A1 and CYP1B1 were significantly higher after PAH4 exposure compared with B[a]P exposure. The results indicated the metabolism of B[a]P was accelerated after PAH4 exposure which might be partly due to the induction of CYPs. These results confirmed PAHs are rapidly metabolized and suggested potential interactions of PAHs may happen among PAH4 mixture.

2.
Food Chem Toxicol ; 177: 113820, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37172713

RESUMO

Mercury is one heavy metal toxin that could cause severe health impairments. Mercury exposure has become a global environmental issue. Mercury chloride (HgCl2) is one of mercury's main chemical forms, but it lacks detailed hepatotoxicity data. The present study aimed to investigate the mechanism of hepatotoxicity induced by HgCl2 through proteomics and network toxicology at the animal and cellular levels. HgCl2 showed apparent hepatotoxicity after being administrated with C57BL/6 mice (16 mg/kg.bw, oral once a day, 28 days) and HepG2 cells (100 µmol/L, 12 h). Otherwise, oxidative stress, mitochondrial dysfunction and inflammatory infiltration play an important role in HgCl2-induced hepatotoxicity. The differentially expressed proteins (DEPs) after HgCl2 treatment and enriched pathways were obtained through proteomics and network toxicology. Western blot and qRT-PCR results showed acyl-CoA thioesterase 1 (ACOT1), acyl-CoA synthetase short chain family member 3 (ACSS3), epidermal growth factor receptor (EGFR), apolipoprotein B (APOB), signal transducer and activator of transcription 3 (STAT3), alanine--glyoxylate aminotransferase (AGXT), cytochrome P450 3A5 (CYP3A5), CYP2E1 and CYP1A2 may be the major biomarkers for HgCl2-induced hepatotoxicity, which involved chemical carcinogenesis, fatty acid metabolism, CYPs-mediated metabolism, GSH metabolism and others. Therefore, this study can provide scientific evidence for the biomarkers and mechanism of HgCl2-induced hepatotoxicity.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Mercúrio , Camundongos , Animais , Humanos , Cloreto de Mercúrio/toxicidade , Cloretos , Células Hep G2 , Proteômica , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Biomarcadores/metabolismo
3.
Food Funct ; 13(23): 12170-12181, 2022 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-36326424

RESUMO

Hepatic steatosis is a common pathological change of liver that manifests as abnormal lipid accumulation. Epidemiological findings support that diseases such as obesity, diabetes, and hyperlipidemia are mostly accompanied by the development of hepatic steatosis. By screening the disease targets of several traditional Chinese medicines (TCMs) with lipid-reducing effects (hawthorn, semen cassiae, etc.) through the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP), we found that peroxisome-activated receptor gamma (PPAR-γ) is involved in regulating several lipid metabolism-related signaling pathways. Further experiments confirmed that PPAR-γ was correlated with aggravated endoplasmic reticulum (ER) stress in overnutrition-induced hepatic steatosis. The stimulation of hepatocytes by abnormal lipid metabolism signals causes an imbalance in ER homeostasis, which subsequently exacerbates hepatocyte lipid abnormalities. The inhibition of glucose regulatory protein 78 (GRP78, a master regulator of ER homeostasis) was effective in reducing hepatocyte PPAR-γ and lipid synthesis levels. In fact, the hawthorn/semen cassiae treatment effectively downregulated hepatocyte ER stress in high-fat-diet fed rats and reduced the PPAR-γ expression as well as related lipid synthesis. Herein, we confirmed that TCMs characterized by natural lipid-lowering effectively target hepatic PPAR-γ and GRP78, improve ER stress, and have a protective effect against obesity-related hepatic steatosis.


Assuntos
Crataegus , Fígado Gorduroso , Ratos , Animais , Dieta Hiperlipídica/efeitos adversos , Estresse do Retículo Endoplasmático , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Sementes/metabolismo , Fígado Gorduroso/metabolismo , Fígado/metabolismo , Obesidade/metabolismo , Lipídeos/farmacologia , Metabolismo dos Lipídeos
4.
FEBS J ; 289(8): 2013-2024, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-33460497

RESUMO

Neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD), are characterized by progressive memory loss and motor impairment. Aging is a major risk factor for neurodegenerative diseases. Neurodegenerative diseases and aging often develop in an irreversible manner and cause a significant socioeconomic burden. When considering their pathogenesis, many studies usually focus on mitochondrial dysfunction and DNA damage. More recently, neuroinflammation, autophagy dysregulation, and SIRT1 inactivation were shown to be involved in the pathogenesis of neurodegenerative diseases and aging. In addition, studies uncovered the role of poly (ADP-ribose)-polymerase-1 (PARP1) in neurodegenerative diseases and aging. PARP1 links to a cluster of stress signals, including those originated by inflammation and autophagy dysregulation. In this review, we summarized the recent research progresses on PARP1 in neurodegenerative diseases and aging, with an emphasis on the relationship among PARP1, neuroinflammation, mitochondria, and autophagy. We discussed the possibilities of treating neurodegenerative diseases and aging through targeting PARP1.


Assuntos
Doenças Neurodegenerativas , Envelhecimento/genética , Autofagia/genética , Humanos , Mitocôndrias/genética , Doenças Neurodegenerativas/genética , Poli(ADP-Ribose) Polimerase-1/genética , Sirtuína 1
5.
J Neuroinflammation ; 18(1): 295, 2021 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-34930303

RESUMO

BACKGROUND: Parkinson's disease (PD) is characterized by degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc), accompanied by accumulation of α-synuclein, chronic neuroinflammation and autophagy dysfunction. Previous studies suggested that misfolded α-synuclein induces the inflammatory response and autophagy dysfunction in microglial cells. The NLRP3 inflammasome signaling pathway plays a crucial role in the neuroinflammatory process in the central nervous system. However, the relationship between autophagy deficiency and NLRP3 activation induced by α-synuclein accumulation is not well understood. METHODS: Through immunoblotting, immunocytochemistry, immunofluorescence, flow cytometry, ELISA and behavioral tests, we investigated the role of p38-TFEB-NLRP3 signaling pathways on neuroinflammation in the α-synuclein A53T PD models. RESULTS: Our results showed that increased protein levels of NLRP3, ASC, and caspase-1 in the α-synuclein A53T PD models. P38 is activated by overexpression of α-synuclein A53T mutant, which inhibited the master transcriptional activator of autophagy TFEB. And we found that NLRP3 was degraded by chaperone-mediated autophagy (CMA) in microglial cells. Furthermore, p38-TFEB pathways inhibited CMA-mediated NLRP3 degradation in Parkinson's disease. Inhibition of p38 had a protective effect on Parkinson's disease model via suppressing the activation of NLRP3 inflammasome pathway. Moreover, both p38 inhibitor SB203580 and NLRP3 inhibitor MCC950 not only prevented neurodegeneration in vivo, but also alleviated movement impairment in α-synuclein A53T-tg mice model of Parkinson's disease. CONCLUSION: Our research reveals p38-TFEB pathways promote microglia activation through inhibiting CMA-mediated NLRP3 degradation in Parkinson's disease, which could be a potential therapeutic strategy for PD. p38-TFEB pathways promote microglia activation through inhibiting CMA-mediated NLRP3 degradation in Parkinson's disease. In this model, p38 activates NLRP3 inflammasome via inhibiting TFEB in microglia. TFEB signaling negatively regulates NLRP3 inflammasome through increasing LAMP2A expression, which binds to NLRP3 and promotes its degradation via chaperone-mediated autophagy (CMA). NLRP3-mediated microglial activation promotes the death of dopaminergic neurons.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Autofagia Mediada por Chaperonas/fisiologia , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Doença de Parkinson/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Autofagia Mediada por Chaperonas/efeitos dos fármacos , Imidazóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Doença de Parkinson/genética , Proteólise/efeitos dos fármacos , Piridinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
6.
Aging Cell ; 19(6): e13163, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32475059

RESUMO

Poly (ADP-ribose) polymerase 1 (PARP1) is a master regulator of diverse biological processes such as DNA repair, oxidative stress, and apoptosis. PARP1 can be activated by aggregated α-synuclein, and this process in turn exacerbates toxicity of α-synuclein. This circle is closely linked to the evolution of Parkinson's disease (PD) that characterized by progressive neurodegeneration and motor deficits. Here, we reported the PARP1, as a novel upstream molecular of transcription factor EB (TFEB), participates in regulation of autophagy in α-synuclein aggregated cells and mice. PARP1 inhibition not only enhances the nuclear transcription of TFEB via SIRT1 mediated down-regulation of mTOR signaling but also reduces nuclear export of TFEB by attenuating the TFEB-CRM1 interaction. Our results revealed that PARP1 inhibition lessened the accumulation of α-synuclein in PD models. Also, oral administration of PARP1 inhibitor Veliparib prevented neurodegeneration and improved motor ability in α-synucleinA53T transgenic mice. These findings identify that PARP1 signaling pathway regulates TFEB-mediated autophagy, pointing to potential therapeutic strategy of PD via enhancing protein degradation systems.


Assuntos
Autofagia/genética , Doença de Parkinson/genética , Poli(ADP-Ribose) Polimerase-1/genética , alfa-Sinucleína/metabolismo , Animais , Humanos , Camundongos , Transfecção
7.
Toxicol Appl Pharmacol ; 388: 114874, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31881179

RESUMO

Abnormal accumulation of α-synuclein and mitochondria dynamics dysfunction are considered to be implicated in the pathogenesis of Parkinson's disease. However, the underlying mechanisms how α-synuclein abnormal accumulation causes mitochondrial dynamics dysfunction remains unclear. Here, we demonstrate that dynamin-related protein 1(DRP1) is a substrate for p38 MAPK, mutant α-synuclein overexpression in SN4741 cell caused p38 MAPK activation, p38 MAPK-mediated phosphorylation DRP1 at serine 616 to activate DRP1 and is associated with increased mitochondrial fission, which resulted in mitochondrial dysfunction and neuronal loss. Inhibition of p38 MAPK or expression of a kinase death form of p38 MAPK not only attenuates DRP1-mediated mitochondrial fission,but also restores the mitochondrial dysfunction and cell death in α-synuclein A53T model. These findings showed that inhibition of p38 MAPK-DRP1 signaling pathway may be a viable therapeutic strategy of PD on maintenance of mitochondrial homeostasis.


Assuntos
Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico , Piridinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Dinaminas/metabolismo , Humanos , Imidazóis/uso terapêutico , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Dinâmica Mitocondrial/genética , Mutação , Neurônios/citologia , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Doença de Parkinson/genética , Doença de Parkinson/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Piridinas/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Serina/metabolismo , alfa-Sinucleína/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Biochem Biophys Res Commun ; 509(2): 390-394, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30594390

RESUMO

Parkinson's disease (PD) is a progressive neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra. Prevailing evidence suggests that abnormal autophagy and mitochondrial dysfunction participate in the process of PD. However, many damages of neuronal functions are regulated by intracellular Ca2+ signaling and the contribution of mitochondrial Ca2+ to the process of neurodegeneration is still unclear. MPP+, the metabolite of a neurotoxin MPTP, causes symptom of PD in animal models by selectively destroying dopaminergic neurons in substantia nigra. Here we report that mitochondrial Ca2+ uniporter (MCU) participated in MPP+-induced autophagic cell death in SH-SY5Y cells. Pharmacological agonist of MCU or exogenous expressed MCU can partially reduce MPP+-induced autophagic cell death. Down-regulation of MCU enhanced autophagic cell death via AMPK activation, which was independent of Beclin1 and PI3K. These findings show that the mitochondrial calcium dyshomeostasis contributes to MPP+-induced neuronal degeneration, and MCU may be a potential therapeutic target of PD through the prevention of pathological autophagy.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/efeitos dos fármacos , Cálcio/metabolismo , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagia/genética , Proteína Beclina-1/genética , Proteína Beclina-1/metabolismo , Biotransformação , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Sinalização do Cálcio , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo
9.
Toxicol Sci ; 165(1): 232-243, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30165626

RESUMO

Impairment in autophagy-lysosomal pathway (ALP) results in accumulation of misfolded proteins and dysfunctional organelles, which is the hallmark of neurodegenerative diseases including Parkinson's disease (PD). Recent studies revealed activated nonreceptor tyrosine kinase Abelson (c-Abl) in PD models and brain specimen of PD patients. Inhibition of c-Abl through pharmacological inhibitors has been shown to enhance ALP function and provide neuroprotective effects in cells and animal models of PD. However, the molecular mechanisms of neuroprotective effects underlying c-Abl inhibition remain elusive. In this study, STI-571, a c-Abl inhibitor, rescued the ALP function through facilitating the nuclear translocation of TFEB and protected against MPP+-induced neuronal cell death. Furthermore, siRNA-mediated knock-down or pharmacological inhibition of GSK3ß mitigated the MPP+-induced neuronal cell death, which was achieved through promoting TFEB nuclear localization and subsequently reversing the function of ALP. Intriguingly, either DPH, c-Abl activator, or MPP+ led to the activation of GSK3ß, which is a negative regulator of TFEB. In addition, c-Abl directly interacted with GSK3ß and catalyzed its phosphorylation at tyrosine 216, and their interaction was enhanced under MPP+ treatment. In contrast, STI-571 abrogated phosphorylation of GSK3ß-Tyr216 induced by MPP+ in SN4741 cells and in primary midbrain neurons. Taken together, these results demonstrate that GSK3ß is a novel c-Abl substrate, and c-Abl-GSk3ß pathway mediates MPP+-induced ALP defects and neuronal cell death, which may represent a potential therapeutic target for PD.


Assuntos
Autofagia/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/metabolismo , Mesilato de Imatinib/farmacologia , Lisossomos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-abl/metabolismo , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Lisossomos/metabolismo , Lisossomos/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Ratos Sprague-Dawley , Transdução de Sinais
10.
Cell Death Dis ; 9(6): 700, 2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29899409

RESUMO

α-synuclein abnormal accumulation and mitochondria dysfunction are involved in the pathogenesis of Parkinson's disease. Selective autophagy of mitochondria (mitophagy) is a crucial component of the network controlling the mitochondrial homeostasis. However, the underlying mechanism that mutant α-synuclein induces mitochondrial abnormality through mitophagy impairment is not fully understood. Here, we showed that mutant A53T α-synuclein accumulation impaired mitochondrial function and Parkin-mediated mitophgy in α-synucleinA53T model. α-synucleinA53T overexpression caused p38 MAPK activation, then p38 MAPK directly phosphorylated Parkin at serine 131 to disrupt the Parkin's protective function. The p38 MAPK inhibition significantly reduced cellular apoptosis, restored mitochondrial membrane potential as well as increased synaptic density both in SN4741 cells and primary midbrain neurons. These findings show that the p38 MAPK-Parkin signaling pathway regulates mitochondrial homeostasis and neuronal degeneration, which may be a potential therapeutic strategy of PD via enhancing mitochondrial turn-over and maintenance.


Assuntos
Apoptose , Mitocôndrias/metabolismo , Proteínas Mutantes/metabolismo , Neurônios/patologia , Doença de Parkinson/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , alfa-Sinucleína/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/ultraestrutura , Mitofagia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Proteínas Quinases/metabolismo , Piridinas/farmacologia , Sinapses/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA