Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Free Radic Biol Med ; 222: 331-343, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38876456

RESUMO

The progressive loss of dopaminergic neurons in the midbrain is the hallmark of Parkinson's disease (PD). A newly emerging form of lytic cell death, ferroptosis, has been implicated in PD. However, it remains unclear in terms of PD-associated ferroptosis underlying causative genes and effective therapeutic approaches. This research explored the underlying mechanism of ferroptosis-related genes in PD. Here, Firstly, we found NOX1 associated with ferroptosis differently in PD patients by bioinformatics analysis. In vitro and in vivo models of PD were constructed to explore the underlying mechanism. qPCR, Western blot analysis, immunohistochemistry, immunofluorescence, Ferro orange, and BODIPY C11 were utilized to analyze the levels of ferroptosis. Transcriptomics sequencing was to investigate the downstream pathway and the analysis of immunoprecipitation to validate the upstream factor. In conclusion, NOX1 upregulation and activation of ferroptosis-related neurodegeneration, therefore, might be useful as a clinical therapeutic agent.


Assuntos
Ferroptose , NADPH Oxidase 1 , Doença de Parkinson , Ferroptose/genética , Humanos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Doença de Parkinson/genética , NADPH Oxidase 1/metabolismo , NADPH Oxidase 1/genética , Animais , Camundongos , Ferritinas/metabolismo , Ferritinas/genética , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Autofagia/genética , Masculino , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
2.
Mol Neurobiol ; 60(10): 6092-6108, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37418066

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disease that affects about 10 million people worldwide. Non-motor and motor symptoms usually accompany PD. Major depressive disorder (MDD) is one of the non-motor manifestations of PD it remains unrecognized and undertreated effectively. MDD in PD has complicated pathophysiologies and remains unclear. The study aimed to explore the candidate genes and molecular mechanisms of PD with MDD. PD (GSE6613) and MDD (GSE98793) gene expression profiles were downloaded from Gene Expression Omnibus (GEO). Above all, the data of the two datasets were standardized separately, and differentially expressed genes (DEGs) were obtained by using the Limma package of R. Take the intersection of the two differential genes and remove the genes with inconsistent expression trends. Subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were investigated to explore the function of the common DEGs. Additionally, the construction of the protein-protein interaction (PPI) network was to search the hub genes, and then the least absolute shrinkage and selection operator (LASSO) regression was used to further identify the key genes. GSE99039 for PD and GSE201332 for MDD were performed to validate the hub genes by the violin plot and receiver operating characteristic (ROC) curve. Last but not least, immune cell dysregulation in PD was investigated by immune cell infiltration. As a result, a total of 45 common genes with the same trend. Functional analysis revealed that they were enriched in neutrophil degranulation, secretory granule membrane, and leukocyte activation. LASSO was performed on 8 candidate hub genes after CytoHubba filtered 14 node genes. Finally, AQP9, SPI1, and RPH3A were validated by GSE99039 and GSE201332. Additionally, the three genes were also detected by the qPCR in vivo model and all increased compared to the control. The co-occurrence of PD and MDD can be attributed to AQP9, SPI1, and RPH3A genes. Neutrophils and monocyte infiltration play important roles in the development of PD and MDD. Novel insights may be gained from the findings for the study of mechanisms.


Assuntos
Transtorno Depressivo Maior , Doenças Neurodegenerativas , Doença de Parkinson , Humanos , Transtorno Depressivo Maior/genética , Doença de Parkinson/genética , Ontologia Genética , Monócitos
3.
Exp Biol Med (Maywood) ; 248(2): 146-156, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36573455

RESUMO

Autophagy plays works by degrading misfolded proteins and dysfunctional organelles and maintains intracellular homeostasis. Apelin-13 has been investigated as an agent that might protect the blood-brain barrier (BBB) from cerebral ischemia/reperfusion (I/R) injury. In this study, we examined whether apelin-13 protects cerebral microvascular endothelial cells, important components of the BBB, from I/R injury by regulating autophagy. To mimic I/R injury, the mouse cerebral microvascular endothelia l cell line bEnd 3 undergoes the process of oxygen and glucose deprivation and re feeding in the process of culture. Cell viability was detected using a commercial kit, and cell migration was monitored by in vitro scratch assay. The tight junction (TJ) proteins ZO-1 and occludin; the autophagy markers LC3 II, beclin 1, and p62; and components of the AKT-mTOR signaling pathway were detected by Western blotting and immunofluorescence. To confirm the role of autophagy in OGD/R and the protective effect of apelin-13, we treated the cells with 3-methyladenine (3-MA), a pharmacological inhibitor of autophagy. Our results demonstrated that OGD/R increased autophagic activity but decreased viability, abundance of TJs, and migration. Viability and TJ abundance were further reduced when the OGD/R group was treated with 3-MA. These results indicated that bEnd.3 upregulates autophagy to ameliorate the effects of OGD/R injury on viability and TJs, but that the autophagy induced by OGD/R alone is not sufficient to protect against the effect on cell migration. Treatment of OGD/R samples with apelin-13 markedly increased viability, TJ abundance, and migration, as well as autophagic activity, whereas 3-MA inhibited this increase, suggesting that apelin-13 exerted its protective effects by upregulating autophagy.


Assuntos
Oxigênio , Traumatismo por Reperfusão , Camundongos , Animais , Oxigênio/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Endoteliais/metabolismo , Glucose/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Autofagia , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/metabolismo , Reperfusão
4.
Biochim Biophys Acta Mol Basis Dis ; 1867(11): 166230, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34358627

RESUMO

Orexin-A (OXA) is a neuropeptide with neuroprotective effect by reducing cerebral ischemia/reperfusion injury (CIRI). Inflammation and apoptosis mediated by astrocyte activation are the key pathological mechanisms for CIRI. We thus attempted to confirm neuroprotective effects of OXA on astrocytic inflammation and apoptosis in CIRI and clarify the relative mechanisms. A middle cerebral artery occlusion and reperfusion (MCAO/R) rat model and U251 glioma cells model subjected to oxygen glucose deprivation and reperfusion (OGD/R) were established, with or without OXA treatment. Neurological deficit score was determined, and cerebral infarct volume was evaluated by 2,3,5-triphenyltetrazolium chloride (TTC) staining. Western Blot was used to detect the expressions of NF-κB p65, p-p65, p-ERK, p-p38, GFAP, OX1R, IL-1ß, TNF-α, IL-6, iNOS, Bcl-2, Bax, CytC, cleaved caspase-9 and cleaved caspase-3 in vivo and in vitro. Pro-inflammatory cytokines in cell supernatant IL-1ß, TNF-α and IL-6 were determined by ELISA. Hoechst 33342 staining was used to detect the apoptosis of astrocyte. Immunofluorescent staining was performed to assess the nuclear translocation of p65 and the expression of GFAP. The results showed that OXA significantly improved neurological deficit score and decreased the volume of infarct area in brain. OXA decreased inflammatory mediators, inhibited astrocyte activation and nuclear translocation of NF-κB and phosphorylation of NF-κB, MAPK/ERK and MAPK/p38. Besides, OXA suppressed apoptosis via upregulating the ratio of Bcl-2/Bax and downregulating cytochrome C, cleaved-caspase-9 and cleaved caspase-3. Overall, it was concluded that OXA exerts neuroprotective effect during CIRI through attenuating astrocytes apoptosis, astrocytes activation and pro-inflammatory cytokines production, by Inhibiting OX1R-mediated NF-κB, MAPK/ERK and MAPK/p38 signaling pathways. The progress in our study is helpful to elucidate the molecular mechanisms of OXA neuroprotection, which could lead to the development of new treatment strategies for ischemic stroke.


Assuntos
Astrócitos/patologia , Infarto da Artéria Cerebral Média/complicações , Orexinas/metabolismo , Traumatismo por Reperfusão/imunologia , Animais , Apoptose/imunologia , Astrócitos/imunologia , Linhagem Celular Tumoral , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/citologia , Córtex Cerebral/imunologia , Córtex Cerebral/patologia , Modelos Animais de Doenças , Humanos , Infarto da Artéria Cerebral Média/imunologia , Infarto da Artéria Cerebral Média/patologia , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , NF-kappa B/metabolismo , Receptores de Orexina/metabolismo , Orexinas/administração & dosagem , Ratos , Traumatismo por Reperfusão/patologia
5.
Neurotox Res ; 39(3): 740-753, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33580874

RESUMO

Oxidative stress and mitochondrial dysfunction are involved in cerebral ischemia/reperfusion injury-induced neuronal apoptosis. Mitophagy is the main method to eliminate dysfunctional mitochondria. Apelin-36, a type of neuropeptide, has been reported to exert protective effects in cerebral I/R (I/R) injury, but its precise mechanisms remain to be elucidated. To study the effects of Apelin-36 on oxidative stress and mitochondrial dysfunction in cerebral I/R injury, the oxygen-glucose deprivation/reperfusion (OGD/R) model with 6 h of ischemia and 6 h of reperfusion was established in HT22 cells. Results demonstrated that Apelin-36 protected against OGD/R injury by improving cell viability, decreasing the apoptotic cells ratio and increasing the ratio of Bcl-2/Bax. In addition, Apelin-36 treatment inhibited oxidative stress by downregulating the level of reactive oxygen species (ROS) and malondialdehyde (MDA) as well as the expression of inducible nitric oxide synthase (iNOS). And Apelin-36 also activated the level of superoxide dismutase (SOD) and glutathione (GSH). Mitochondrial apoptosis was also alleviated with Apelin-36 treatment detected by the mitochondrial membrane potential (MMP) and the expression of Cytochrome c (Cyt c), Cleaved caspase-9, and Cleaved caspase-3. Furthermore, the SIRT1-mediated PINK1/Parkin-dependent mitophagy was activated by Apelin-36 treatment with the downregulation of p62 and upregulation of LC3B-II and Beclin1. Both EX527 and Cyclosporine A (CsA), which are inhibitors of SIRT1 and mitophagy, markedly alleviated the inhibition of oxidative stress and mitochondrial dysfunction caused by Apelin-36. These findings suggest that SIRT1-mediated PINK1/Parkin-dependent mitophagy is involved in the neuroprotective effects of Apelin-36 on OGD/R-induced oxidative stress and mitochondrial dysfunction.


Assuntos
Apelina/farmacologia , Glucose/deficiência , Mitocôndrias/metabolismo , Proteínas Quinases/metabolismo , Sirtuína 1/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Linhagem Celular Transformada , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Mitofagia/fisiologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo
6.
Neuropeptides ; 87: 102134, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33639357

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder, characterized by the loss of dopaminergic neurons in the substantia nigra and the deposition of Lewy bodies. Mitochondrial dysfunction, oxidative stress, and autophagy dysfunction are involved in the pathogenesis of PD. Ghrelin is a brain-gut peptide that has been reported that protected against 1-methyl-4-phenyl-1,2,3,6- tetrahydropyran (MPTP)/MPP+-induced toxic effects. In the present work, human neuroblastoma SH-SY5Y cells were exposed to rotenone as a PD model to explore the underlying mechanism of ghrelin. We found that ghrelin inhibited rotenone-induced cytotoxicity, mitochondrial dysfunction, and apoptosis by improving cell viability, increasing the ratio of red/green of JC-1, inhibiting the production of reactive oxidative species (ROS), and regulating Bcl-2, Bax, Cytochrome c, caspase-9, and caspase-3 expression. Besides, ghrelin promoted mitophagy accompanied by up-regulating microtubule-associated protein 1 Light Chain 3B-II/I(LC3B-II/I) and Beclin1 but decreasing the expression of p62. Moreover, ghrelin promoted PINK1/Parkin mitochondrial translocation. Additionally, we investigated that ghrelin activated the AMPK/SIRT1/PGC1α pathway and pharmacological inhibition of AMPK and SIRT1 abolished the cytoprotection of ghrelin, decreased the level of mitophagy, and PINK1/Parkin mitochondrial translocation. Taken together, our findings suggested that mitophagy and AMPK/SIRT1/PGC1α pathways were related to the cytoprotection of ghrelin. These findings provided novel insights into the underlying mechanisms of ghrelin, further mechanistic studies on preclinical and clinical levels are required to be conducted with ghrelin to avail and foresee it as a potential agent in the treatment and management of PD.


Assuntos
Grelina/fisiologia , Mitocôndrias/efeitos dos fármacos , Mitofagia/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Rotenona/toxicidade , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/fisiologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Grelina/farmacologia , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neuroblastoma , Estresse Oxidativo/efeitos dos fármacos , Doença de Parkinson , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/fisiologia , Proteínas Quinases/metabolismo , Transporte Proteico/efeitos dos fármacos , Espécies Reativas de Oxigênio , Rotenona/antagonistas & inibidores , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , alfa-Sinucleína/biossíntese , alfa-Sinucleína/genética
7.
Mol Med Rep ; 23(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33576468

RESUMO

Orexin­A (OXA) protects neurons against cerebral ischemia­reperfusion injury (CIRI). Endoplasmic reticulum stress (ERS) induces apoptosis after CIRI by activating caspase­12 and the CHOP pathway. The present study aimed to determine whether OXA mitigates CIRI by inhibiting ERS­induced neuronal apoptosis. A model of CIRI was established, in which rats were subjected to middle cerebral artery occlusion with ischemic intervention for 2 h, followed by reperfusion for 24 h. Neurological deficit examination and 2,3,5­triphenyltetrazolium chloride staining were performed to assess the level of CIRI and neuroprotection by OXA. Expression levels of ERS­related proteins and cleaved caspase­3 were measured via western blotting, while the rate of neuronal apoptosis in the cortex was determined using a TUNEL assay. OXA treatment decreased the infarct volume of rats after CIRI and attenuated neuron apoptosis. Furthermore, administration of OXA decreased the expression levels of GRP78, phosphorylated (p)­PERK, p­eukaryotic initiation factor­2α, p­inositol requiring enzyme 1α, p­JNK, cleaved caspase­12, CHOP and cleaved caspase­3, all of which were induced by CIRI. Collectively, these findings suggested that OXA attenuated CIRI by inhibiting ERS­mediated apoptosis, thus clarifying the mechanism underlying its neuroprotective effect and providing a novel therapeutic direction for the treatment of CIRI.


Assuntos
Apoptose/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Orexinas/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Caspase 12/metabolismo , Caspase 3/metabolismo , Proteínas de Choque Térmico/metabolismo , Infarto da Artéria Cerebral Média/complicações , Injeções Intraventriculares , Masculino , Neurônios/citologia , Neurônios/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Orexinas/administração & dosagem , Fosfoproteínas/metabolismo , Ratos Sprague-Dawley , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/fisiopatologia , Fator de Transcrição CHOP/metabolismo
8.
Neuropeptides ; 85: 102112, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33333485

RESUMO

Ghrelin is a regulatory peptide that is the endogenous ligand of the growth hormone secretagogue 1a (GHS-R1a) which belongs to the G protein-coupled receptor family. Ghrelin and GHS-R1a are widely expressed in the central and peripheral tissues and play therapeutic potential roles in the cytoprotection of many internal organs. Endoplasmic reticulum stress (ERS), oxidative stress, and autophagy dysfunction, which are involved in various diseases. In recent years, accumulating evidence has suggested that ghrelin exerts protective effects by regulating ERS, oxidative stress, and autophagy in diverse diseases. This review article summarizes information about the roles of the ghrelin system on ERS, oxidative stress, and autophagy in multiple diseases. It is suggested that ghrelin positively affects the treatment of diseases and may be considered as a therapeutic drug in many illnesses.


Assuntos
Autofagia/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Grelina/metabolismo , Grelina/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Animais , Humanos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transdução de Sinais
9.
Neural Regen Res ; 16(6): 1044-1051, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33269749

RESUMO

Apelin-13 is a novel endogenous ligand for an angiotensin-like orphan G-protein coupled receptor, and it may be neuroprotective against cerebral ischemia injury. However, the precise mechanisms of the effects of apelin-13 remain to be elucidated. To investigate the effects of apelin-13 on apoptosis and autophagy in models of cerebral ischemia/reperfusion injury, a rat model was established by middle cerebral artery occlusion. Apelin-13 (50 µg/kg) was injected into the right ventricle as a treatment. In addition, an SH-SY5Y cell model was established by oxygen-glucose deprivation/reperfusion, with cells first cultured in sugar-free medium with 95% N2 and 5% CO2 for 4 hours and then cultured in a normal environment with sugar-containing medium for 5 hours. This SH-SY5Y cell model was treated with 10-7 M apelin-13 for 5 hours. Results showed that apelin-13 protected against cerebral ischemia/reperfusion injury. Apelin-13 treatment alleviated neuronal apoptosis by increasing the ratio of Bcl-2/Bax and significantly decreasing cleaved caspase-3 expression. In addition, apelin-13 significantly inhibited excessive autophagy by regulating the expression of LC3B, p62, and Beclin1. Furthermore, the expression of Bcl-2 and the phosphatidylinositol-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway was markedly increased. Both LY294002 (20 µM) and rapamycin (500 nM), which are inhibitors of the PI3K/Akt/mTOR pathway, significantly attenuated the inhibition of autophagy and apoptosis caused by apelin-13. In conclusion, the findings of the present study suggest that Bcl-2 upregulation and mTOR signaling pathway activation lead to the inhibition of apoptosis and excessive autophagy. These effects are involved in apelin-13-induced neuroprotection against cerebral ischemia/reperfusion injury, both in vivo and in vitro. The study was approved by the Animal Ethical and Welfare Committee of Jining Medical University, China (approval No. 2018-JS-001) in February 2018.

10.
Cell Signal ; 79: 109839, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33212156

RESUMO

Orexin A (OXA) is a neuroprotective peptide that exerts protective effects on multiple physiological and pathological processes. Activation of autophagy is linked to the occurrence of cerebral ischemia-reperfusion injury (CIRI); however, its function remains incompletely understood. In this study, OXA was sought to exert its neuroprotective role by regulating autophagy in oxygen and glucose deprivation and reoxygenation (OGD/R) model and middle cerebral artery occlusion (MCAO) model of rats, and to elucidate the underlying molecular mechanisms. Acridine orange (AO) staining was used to evaluate autophagic vacuoles. Cell viability was measured by CCK8. The levels of p-ERK1/2, t-ERK1/2, p-mTOR, LC3B, Beclin 1, and p62 were evaluated by western blotting. Apoptosis rate was detected by Hoechst 33342 staining and Terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling (TUNEL). OXA treatment alleviated neuronal apoptosis and significantly inhibited autophagy activity. Mechanistically, OXA exerted its neuroprotective effects in vivo and in vitro by suppressing over-activated autophagy by modulating OX1R-mediated MAPK/ERK/mTOR pathway. The results of this study elucidate the roles of autophagy in CIRI and the mechanisms underlying the neuroprotective action of OXA. Our findings could facilitate the development of novel therapeutics for ischemic stroke.


Assuntos
Autofagia , Transtornos Cerebrovasculares , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Traumatismo por Reperfusão , Serina-Treonina Quinases TOR/metabolismo , Animais , Linhagem Celular Tumoral , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/prevenção & controle , Humanos , Masculino , Ratos , Ratos Wistar , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle
11.
Peptides ; 133: 170374, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32814076

RESUMO

Parkinson's disease (PD) is a common progressive and multifactorial neurodegenerative disease. Current pharmacological therapies for PD are inadequate and often accompanied by serious side effects. In search of neuroprotective agents being considered to be beneficial to PD therapy. Ghrelin confers neuroprotective effect in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned PD model, but the underlying mechanism remains not fully elucidated. Here, we utilized human neuroblastoma SH-SY5Y cells exposed to MPP+ as a PD model to investigate the underlying mechanism of Ghrelin. In our present work, cell viability, cell apoptosis, oxidative stress-related indicators, and the level of Nrf2, HO-1, PERK, eIF2α, ATF4, CHOP, and ERK1/2 were examined. The results showed that Ghrelin attenuated MPP+-induced change of cell viability, apoptosis, coupled with decreased Cytochrome c, caspase-9, and caspase-3 expressions. Consistently, Ghrelin suppressed MPP+-induced oxidative stress. Moreover, Ghrelin markedly enhanced Nrf2 expression and nuclear accumulation as well as HO-1 induction. Further investigations showed that Ghrelin significantly inhibited the endoplasmic reticulum stress PERK-eIF2α-ATF4-CHOP pathway. Interestingly, we then found that Ghrelin promoted phosphorylation of ERK1/2, and pharmacological inhibition of ERK signaling abolished the cytoprotective effect of Ghrelin. Furthermore, we also found promoting the activation of the Nrf2/ HO-1 pathway and suppressing of the PERK pathway were mediated by ERK1/2. These findings provided novel insights into the underlying mechanisms of Ghrelin exerted protective effect, suggesting its potential as a novel therapeutic strategy against PD.


Assuntos
1-Metil-4-fenilpiridínio/toxicidade , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Grelina/farmacologia , Doença de Parkinson/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Grelina/metabolismo , Heme Oxigenase-1/metabolismo , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , eIF-2 Quinase/metabolismo
12.
Brain Res ; 1746: 147023, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32710901

RESUMO

Parkinson's disease (PD) is the second most common progressive neurodegenerative disorder, the important pathology of PD due to the prominent loss of the dopaminergic neurodegeneration in the substantia nigra pars compacta (SNpc) and striatum (STR). Although the etiology of PD is not fully understood, aggregation of α-synuclein, impaired autophagy, and endoplasmic reticulum stress (ERS) are involved in the pathogenesis of PD. Previously it has been demonstrated that Ghrelin is a kind of peptide protected dopaminergic neurons against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyran (MPTP)-induced neurotoxicity, but the detailed mechanism remains to be elucidated. In the present work, we investigated the effects of Ghrelin on autophagy and ERS-mediated apoptosis in the MPTP-lesioned PD mice model. We found that Ghrelin was neuroprotective against MPTP-induced dopaminergic neurodegeneration. Subsequently, we investigated Ghrelin inhibited the accumulation and phosphorylation of α-synuclein induced by MPTP. Moreover, Ghrelin promoted autophagy indicated by the up-regulation of microtubule-associated protein 1 Light Chain 3B-II/I (LC3B-II/I) and Beclin1, as well as decreasing the level of p62 in the SNpc and STR. Besides, the activation of the ERS-related apoptosis signaling pathway including IRE1α and Caspase-12 signaling pathway induced by MPTP was suppressed by Ghrelin treatment. Furthermore, Ghrelin also decreased Caspase-3 expression. Taken together, our results indicated that Ghrelin may exert neuroprotective effects via regulating α-synuclein activities, enhancing autophagy, and ameliorating ERS-mediated apoptosis in MPTP-lesioned mice, which provides a new target for potential pharmacologic interventions of PD treatment in the future.


Assuntos
Encéfalo/patologia , Neurônios Dopaminérgicos/patologia , Grelina/farmacologia , Intoxicação por MPTP/patologia , Fármacos Neuroprotetores/farmacologia , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
13.
Cell Signal ; 73: 109671, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32407761

RESUMO

Apelin receptor (APJ) and bradykinin B2 receptor (B2R) play an important role in many physiological processes and share multiple similar characteristics in distribution and functions in the cardiovascular system. We first identified the endogenous expression of APJ and B2R in human umbilical vein endothelial cells (HUVECs) and their co-localization on human embryonic kidney (HEK) 293 cells membrane. A suite of bioluminescence and fluorescence resonance energy transfer (BRET and FRET), proximity ligation assay (PLA), and co-immunoprecipitation (Co-IP) was exploited to demonstrate formation of functional APJ and B2R heterodimer in HUVECs and transfected cells. Stimulation with apelin-13 and bradykinin (BK) increased the phosphorylation of the endothelial nitric oxide synthase (eNOS) in HUVECs, which could be inhibited by the silencing of APJ or B2R, indicating the APJ-B2R dimer is critical for eNOS phosphorylation in HUVECs. Furthermore, the increase of NOS and extracellular signal regulated kinases1/2 (ERK1/2) phosphorylation mediated by APJ/B2R dimer can be inhibited by U0126 and U73122, respectively, suggesting that the heterodimer might activate the PLC/ERK1/2/eNOS signaling pathway, and finally leading to a significant increase in cell proliferation. Thus, we uncovered for the first time the existence of APJ-B2R heterodimer and provided a promising new target in cardiovascular therapeutics.


Assuntos
Receptores de Apelina/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Receptor B2 da Bradicinina/metabolismo , Proliferação de Células , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Sistema de Sinalização das MAP Quinases , Multimerização Proteica
14.
Brain Res ; 1726: 146493, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31586624

RESUMO

Parkinson's disease (PD), a common human neurodegenerative disorder, is characterized by the presence of intraneuronal Lewy bodies composed principally of abnormal aggregated and post-translationally modified α-synuclein. In our previous research, we have demonstrated the neuroprotective effect of Apelin-36, a neuroendocrine peptide in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP)-lesioned PD model mice. Therefore, this study was designed to evaluate the neuroprotective mechanism of Apelin-36 against MPTP-induced neurotoxicity in mice. The results showed that MPTP-induced the depletion of dopamine in the striatum (STR) was partially reversed by Apelin-36. Apelin-36 also improved the activity of antioxidant system including superoxide dismutase (SOD) and glutathione (GSH), and decreased the overproduction of malondialdehyde (MDA) in the substantia nigra pars compacta (SNpc) and STR of MPTP-treated mice. Moreover, Apelin-36 downregulated inducible nitric oxide synthase (iNOS) and nitrated α-synuclein expression. Furthermore, Apelin-36 significantly promoted autophagy indicated by the up-regulation of LC3-II and Beclin1 and inhibition of p62 expression in the SNpc and STR of MPTP-treated mice. The protective effect of Apelin-36 was also associated with the inhibition of the apoptosis signal-regulating kinase 1 (ASK1)/c-Jun N-terminal kinase (JNK) signaling pathway and inactivation of caspase-3. Taken together, our findings demonstrated that the neuroprotective mechanism of Apelin-36 against MPTP-induced neurotoxicity in mice might be related to decreasing the aggregation of nitrated α-synuclein and alleviating oxidative stress as well as promoting autophagy and inhibiting ASK1/JNK/caspase-3 apoptotic pathway, which provides a novel strategy for PD treatment.


Assuntos
Apelina/administração & dosagem , Apelina/metabolismo , Autofagia , Intoxicação por MPTP/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Estresse Oxidativo , Animais , Autofagia/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Parte Compacta da Substância Negra/efeitos dos fármacos , Parte Compacta da Substância Negra/metabolismo , Transdução de Sinais/efeitos dos fármacos
15.
Front Neurol ; 11: 608610, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33424757

RESUMO

Mitochondria are important places for eukaryotes to carry out energy metabolism and participate in the processes of cell differentiation, cell information transmission, and cell apoptosis. Autophagy is a programmed intracellular degradation process. Mitophagy, as a selective autophagy, is an evolutionarily conserved cellular process to eliminate dysfunctional or redundant mitochondria, thereby fine-tuning the number of mitochondria and maintaining energy metabolism. Many stimuli could activate mitophagy to regulate related physiological processes, which could ultimately reduce or aggravate the damage caused by stimulation. Stroke is a common disease that seriously affects the health and lives of people around the world, and ischemic stroke, which is caused by cerebral vascular stenosis or obstruction, accounts for the vast majority of stroke. Abnormal mitophagy is closely related to the occurrence, development and pathological mechanism of ischemic stroke. However, the exact mechanism of mitophagy involved in ischemic stroke has not been fully elucidated. In this review, we discuss the process and signal pathways of mitophagy, the potential role of mitophagy in ischemic stroke and the possible signal transduction pathways. It will help deepen the understanding of mitophagy and provide new ideas for the treatment of ischemic stroke.

16.
Front Physiol ; 10: 1038, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31474876

RESUMO

Extracellular signal-regulated kinase 1/2 (ERK1/2), an important member of the mitogen-activated protein kinase family, is found in many organisms, and it participates in intracellular signal transduction. Various stimuli induce phosphorylation of ERK1/2 in vivo and in vitro. Phosphorylated ERK1/2 moves to the nucleus, activates many transcription factors, regulates gene expression, and controls various physiological processes, finally inducing repair processes or cell death. With the aging of the population around the world, the occurrence of ischemia-reperfusion injury (IRI), especially in the brain, heart, kidney, and other important organs, is becoming increasingly serious. Abnormal activation of the ERK1/2 signaling pathway is closely related to the development and the metabolic mechanisms of IRI. However, the effects of this signaling pathway and the underlying mechanism differ between various models of IRI. This review summarizes the ERK1/2 signaling pathway and the molecular mechanism underlying its role in models of IRI in the brain, heart, liver, kidneys, and other organs. This information will help to deepen the understanding of ERK1/2 signals and deepen the exploration of IRI treatment based on the ERK1/2 study.

17.
Brain Res ; 1721: 146334, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31306618

RESUMO

Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons within the substantia nigra compacta (SNpc) which leads to the behavioral dysfunction. In the present study, we investigated the effect of Apelin-36 on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP)/1-methyl-4-phenylpyridinium (MPP+)-induced neurotoxicity. The treatment with Apelin-36 significantly alleviated the MPTP-induced the behavioral dysfunction and dopaminergic neurodegeneration in the SNpc of mice, and also remarkably decreased the MPP+-induced cell death of SH-SY5Y cells. Furthermore, Apelin-36 reversed the MPTP/MPP+-induced loss of TH expression and the induction of α-synuclein expression. Additionally, Apelin-36 significantly attenuated the endoplasmic reticulum stress (ERS) indicated by the inhibition of GRP78, CHOP and cleaved caspase-12 expression in MPTP/MPP+ treated mice and cells. Taken together, the results indicated that Apelin-36 attenuates MPTP/MPP+-induced neurotoxicity, and suggested that Apelin-36 could be a potential therapeutic strategy for the treatment of PD.


Assuntos
Apelina/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , alfa-Sinucleína/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Apelina/fisiologia , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Intoxicação por MPTP/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/fisiopatologia
18.
Cell Signal ; 62: 109348, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31233841

RESUMO

The neuropeptide orexin-A (OXA) has a neuroprotective effect, acting as an anti-apoptotic factor in response to multiple stimuli. Apoptosis induced by endoplasmic reticulum stress (ERS) underlies oxygen-glucose deprivation and reoxygenation (OGD/R)-induced cell damage, an in vitro model of ischemia/reperfusion injury. However, that OXA inhibits ERS-induced apoptosis in the OGD/R model has not been reported. In the present study, we investigated the neuroprotective effect of OXA (0.1 µM) on OGD/R-induced damage in the human neuroblastoma cell line SH-SY5Y. After OXA treatment following 4 h oxygen-glucose deprivation (OGD) and then 4 h reoxygenation (R), cell morphology, viability, and apoptosis were analyzed by histology, Cell Counting Kit-8 assay, and flow cytometry, respectively. Western blotting was used to measure expression levels of ERS- and apoptosis-related proteins. To determine signaling pathways involved in OXA-mediated neuroprotection, the Gi pathway inhibitor pertussis toxin (PTX; 100 ng/mL) and PI3K inhibitor LY294002 (LY; 10 µM) were added. In addition, in order to prove the specificity of these characteristics, the OXA antagonist Suvorexant (DORA; Ki of 0.55 nM and 0.35 nM for OX1R and OX2R) was used for intervention. Our results showed that OGD/R induced cell damage, manifested as morphological changes and a significant decrease in viability. Furthermore, Western blotting detected an increase in ERS-related proteins GRP78, p-IRE1α, p-JNK, and Cleaved caspase-12, as well as apoptosis-related proteins Cleaved caspase-3 and Bax, and a decrease in the anti-apoptosis factor Bcl-2. OXA intervention alleviated the degree of cellular damage, and protein expression was also reversed. In addition, the protective effect of OXA was reduced by adding PTX and LY. Meanwhile, after the use of DORA, changes in the expression of related proteins were detected, and it was found that the protective effect of OXA was weakened. Collectively, our results indicate that OXA has a neuroprotective effect on OGD/R-induced cell damage by inhibiting ERS-induced apoptosis through the combined action of Gi and PI3K signaling pathways. These findings help to clarify the mechanism underlying the neuroprotective action of OXA, which should aid the development of further candidate drugs, and provide a new therapeutic direction for the treatment of ischemic stroke.


Assuntos
Apoptose/efeitos dos fármacos , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Orexinas/genética , Traumatismo por Reperfusão/genética , Cromonas/farmacologia , Dano ao DNA/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/genética , Glucose/efeitos adversos , Humanos , Morfolinas/farmacologia , Fármacos Neuroprotetores/farmacologia , Orexinas/farmacologia , Oxigênio/efeitos adversos , Fosfatidilinositol 3-Quinases/genética , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos
19.
Sheng Li Xue Bao ; 71(3): 454-462, 2019 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-31218336

RESUMO

Neural stem cell therapy, as a new therapeutic method for neural diseases, has aroused a wide concern for over 20 years since neural stem cells were first found in 1992. Ischemic stroke is highly concerned because of its high incidence, mortality and disability rates. Because the brain has a limited ability to repair itself, to improve neural function and promote neural regeneration may help to prevent occurrence and development of neurological diseases. It is noteworthy that some stroke patients showed an ability to repair brain several months after the stroke happened, suggesting an existence of endogenous nerve repair in these patients. The research advances in functions of endogenous neural stem cells in neural regeneration and the related regulators after ischemic stroke are summarized in this review to provide new views of the mechanism of neural functional recovery after ischemic stroke.


Assuntos
Isquemia Encefálica/terapia , Regeneração Nervosa , Células-Tronco Neurais/citologia , Acidente Vascular Cerebral/terapia , Humanos
20.
Brain Res ; 1715: 203-212, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-30914252

RESUMO

The dopaminergic neurodegeneration in the substantia nigrapars compacta (SNpc) and striatum of the midbrain is the important pathological feature of Parkinson's disease (PD). It has been shown that autophagy and endoplasmic reticulum stress (ERS) are involved in the occurrence and development of PD. The neuropeptide Apelin-13 is neuroprotective in the neurological diseases such as PD, Alzheimer's disease and cerebral ischemic stroke. In the present work, we investigated the neuroprotective effects of Apelin-13 on ERS and autophagy in the dopaminergic neurodegeneration of SNpc of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP)-treated mice. The intranigral injection of Apelin-13 alleviated the behavioral dysfunction and dopaminergic neurodegeneration induced by MPTP. After the exposure to MPTP, the expression of tyrosine hydroxylase (TH) was significantly decreased as well as the increased α-synuclein expression, which was significantly reversed by the intranigral injection of Apelin-13. Also, Apelin-13 significantly reversed the decreasing autophagy induced by MPTP which was indicated by the up-regulation of LC3B-II and Beclin1 and down-regulation of p62. And MPTP-induced ERS such as IRE1α, XBP1s, CHOP and GRP78 was significantly inhibited by Apelin-13. Taken together, Apelin-13 protects dopaminergic neurons in MPTP-induced PD model mice in vivo through inhibiting ERS and promoting autophagy, which contributes to the therapy for PD in the future.


Assuntos
Autofagia/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Apelina/metabolismo , Apelina/farmacologia , Proteína Beclina-1/metabolismo , Encéfalo/metabolismo , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/metabolismo , Substância Negra/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA