Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Aging Neurosci ; 12: 78, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32296327

RESUMO

AIM: To investigate the role of histone deacetylase 6 (HDAC6) deacetylation activity in nucleotide-binding oligomerization domain and leucine-rich repeat pyrin 3 domain (NLRP3) inflammatory response and explore the effects of pharmacological inhibition of HDAC6 with tubastatin A (TBA) on dopaminergic injury. METHODS: Using 6-OHDA-induced Parkinson's disease (PD) models, we examined the effects of TBA on NLRP3 activation and cell injury in SH-SY5Y cells. We also investigated the effects of TBA on NLRP3 inflammatory responses and dopaminergic injury in the nigrostriatal system in mice and analyzed the acetylation levels of peroxiredoxin2 (Prx2) and oxidative stress. RESULTS: TBA inhibited 6-OHDA-induced NLRP3 activation, as demonstrated by decreased expressions of NLRP3 and matured caspase-1 and IL-1ß, and also alleviated glial proliferation and dopaminergic neuronal degeneration. Notably, TBA recovered acetylation levels of Prx2 and reduced oxidative stress. CONCLUSION: Our findings indicate that pharmacological inhibition of HDAC6 with TBA attenuates NLRP3 inflammation and protects dopaminergic neurons, probably through Prx2 acetylation. This study suggests that the deacetylase catalytic domain of HDAC6 is a potential target for PD treatment.

2.
FASEB J ; 33(11): 12164-12174, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31370707

RESUMO

Variants in the gene encoding the triggering receptor expressed on myeloid cells 2 (TREM2) are known to increase the risk of developing Alzheimer disease and Parkinson's disease (PD). However, the potential role of TREM2 effect on synucleinopathy has not been characterized. In this study, we investigated whether loss of TREM2 function affects α-synucleinopathy both in vitro and in vivo. In vitro, BV2 microglial cells were exposed to α-synuclein (α-syn) in the presence or absence of TREM2 small interference RNA. For in vivo studies, wild-type controls and TREM2 gene knockout mice were intracranially injected in the substantia nigra with adeno-associated viral vectors expressing human α-syn (AAV-SYN) to induce PD. Our results revealed that knockdown of TREM2 aggravated α-syn-induced inflammatory responses in BV2 cells and caused greater apoptosis in SH-SY5Y cells treated with BV2-conditioned medium. In mice, TREM2 knockout exacerbated dopaminergic neuron loss in response to AAV-SYN. Moreover, both in vitro and in vivo TREM2 deficiency induced a shift from an anti-inflammatory toward a proinflammatory activation status of microglia. These data suggest that impairing microglial TREM2 signaling aggravates proinflammatory responses to α-syn and exacerbates α-syn-induced neurodegeneration by modulating microglial activation state.-Guo, Y., Wei, X., Yan, H., Qin, Y., Yan, S., Liu, J., Zhao, Y., Jiang, F., Lou, H. TREM2 deficiency aggravates α-synuclein-induced neurodegeneration and neuroinflammation in Parkinson's disease models.


Assuntos
Inflamação/etiologia , Glicoproteínas de Membrana/fisiologia , Microglia/fisiologia , Doença de Parkinson/etiologia , Receptores Imunológicos/fisiologia , alfa-Sinucleína/farmacologia , Animais , Apoptose , Células Cultivadas , Modelos Animais de Doenças , Feminino , Masculino , Glicoproteínas de Membrana/deficiência , Camundongos , Microglia/efeitos dos fármacos , Microglia/patologia , Receptores Imunológicos/deficiência , Transdução de Sinais
3.
FASEB J ; 33(5): 6584-6595, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30794438

RESUMO

LPS has been shown to elicit neuroinflammation associated with the up-regulation of the eicosanoid pathway in animal models; however, the regulatory mechanisms of TLR4 in brain neuroinflammatory conditions remain elusive. ß-Arrestins are key regulators of the GPCR signaling pathway and are involved in the leukotriene B4-induced leukocyte migration to initiate inflammatory response. However, the roles of ß-arrestins in eicosanoid regulation and related diseases are not clear. To address this issue, we conducted a study to investigate the effect of TLR4 on the eicosanoid pathway in ischemic stroke brain and to explore the underlying molecular regulation mechanism. Cerebral ischemia was produced by occlusion of the middle cerebral artery, followed by reperfusion for 24 h. We demonstrated that knockout of TLR4 improves ischemic stroke brain associated with eicosanoid down-regulation. Interestingly, genetic disruption of ß-arrestin-2 failed to decrease neuroinflammation in the damaged brain of TLR4-/- mice, which indicates the requirement of ß-arrestin-2 for TLR4 knockdown protection. Further study showed that the negative regulation of phosphorylated (phospho-)ERK1/2 and phospho-cytosolic phospholipase A2 α (cPLA2α) by TLR4 deficiency was eliminated by genetic disruption of ß-arrestin-2. In addition, ß-arrestin-2 deficiency reversed the reduction of colocalization of phospho-ERK1/2 with phospho-cPLA2α in TLR4-/- mice following ischemic stroke. Mechanistic studies indicated that ß-arrestin-2 specifically colocalized and associated with ERK1/2 to prevent ERK1/2-dependent cPLA2α activation following ischemic injury, and ß-arrestin-2 deficiency blocked the negative regulation of phospho-ERK1/2, revived the association of phospho-ERK1/2 with phospho-cPLA2α, and subsequently increased the prostaglandin E2 and thromboxane A2 production remarkably. Our findings may provide novel insights that ß-arrestin-2 is responsible for ischemic brain improvement in TLR4-/- mice via negative regulation of eicosanoid production.-Xiang, Y., Wei, X., Du, P., Zhao, H., Liu, A., Chen, Y. ß-Arrestin-2-ERK1/2 cPLA2α axis mediates TLR4 signaling to influence eicosanoid induction in ischemic brain.


Assuntos
Isquemia Encefálica/metabolismo , Dinoprostona/biossíntese , Fosfolipases A2 do Grupo IV/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Tromboxano A2/biossíntese , Receptor 4 Toll-Like/metabolismo , beta-Arrestina 2/metabolismo , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Dinoprostona/genética , Fosfolipases A2 do Grupo IV/genética , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Tromboxano A2/genética , Receptor 4 Toll-Like/genética , beta-Arrestina 2/genética
4.
J Cell Mol Med ; 23(5): 3325-3335, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30793522

RESUMO

We previously reported that nucleotide-binding oligomerization domain-containing protein (NOD) 2 was involved in the inflammatory responses to cerebral ischaemia/reperfusion (I/R) insult. However, the mechanism by which NOD2 participates in brain ischaemic injury and the regulation of NOD2 in the process are still obscure. Increased ß-arrestin 2 (ARRB2) expression was observed in microglia following cerebral I/R in wild-type mice besides the up-regulation of NOD2 and TRAF6. Stimulation of NOD2 by muramyl dipeptide (MDP) in BV2 cells induced the activation of NF-κB by the phosphorylation of p65 subunit and the degradation of IκBα. Meanwhile, the protein level of Cyclooxygenase-2 (COX-2), the protein expression and activity of MMP-9 were significantly increased in BV2 cells after administration of MDP. Furthermore, overexpression of ARRB2 significantly suppressed the inflammation induced by MDP, silence of ARRB2 significantly enhanced the inflammation induced by MDP in BV2 cells. In addition, we observed endogenous interaction of TRAF6 and ARRB2 after stimulation of MDP or cerebral I/R insult, indicating ARRB2 negatively regulates NOD2-triggered inflammatory signalling pathway by associating with TRAF6 in microglia after cerebral I/R injury. Finally, the in vivo study clearly confirmed that ARRB2 negatively regulated NOD2-induced inflammatory response, as ARRB2 deficiency exacerbated stroke outcomes and aggravated the NF-κB signalling pathway induced by NOD2 stimulation after cerebral I/R injury. These findings revealed ARRB2 negatively regulated NOD2 signalling pathway through the association with TRAF6 in cerebral I/R injury.


Assuntos
Isquemia Encefálica/metabolismo , Microglia/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Traumatismo por Reperfusão/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , beta-Arrestina 2/metabolismo , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Animais , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Ciclo-Oxigenase 2/metabolismo , Inflamação/patologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia
5.
J Immunol ; 202(3): 747-759, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30598514

RESUMO

G protein-coupled receptor 120 (GPR120) has been shown to negatively regulate inflammation and apoptosis, but its role in cerebral ischemic injury remains unclear. Using an in vivo model of middle cerebral artery occlusion (MCAO) and an in vitro model of oxygen-glucose deprivation (OGD), we investigated the potential role and molecular mechanisms of GPR120 in focal cerebral ischemic injury. Increased GPR120 expression was observed in microglia and neurons following MCAO-induced ischemia in wild type C57BL/6 mice. Treatment with docosahexaenoic acid (DHA) inhibited OGD-induced inflammatory response in primary microglia and murine microglial BV2 cells, whereas silencing of GPR120 strongly exacerbated the inflammation induced by OGD and abolished the anti-inflammatory effects of DHA. Mechanistically, DHA inhibited OGD-induced inflammation through GPR120 interacting with ß-arrestin2. In addition to its anti-inflammatory function, GPR120 also played a role in apoptosis as its knockdown impaired the antiapoptotic effect of DHA in OGD-induced rat pheochromocytoma (PC12) cells. Finally, using MCAO mouse model, we demonstrated that GPR120 activation protected against focal cerebral ischemic injury by preventing inflammation and apoptosis. Our study indicated that pharmacological targeting of GPR120 may provide a novel approach for the treatment of patients with ischemic stroke.


Assuntos
Apoptose , Isquemia Encefálica/prevenção & controle , Inflamação/prevenção & controle , Receptores Acoplados a Proteínas G/metabolismo , Ativação Transcricional , Animais , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/farmacologia , Inativação Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Células PC12 , Ratos , Receptores Acoplados a Proteínas G/genética , Acidente Vascular Cerebral/prevenção & controle
6.
J Neuroinflammation ; 15(1): 243, 2018 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-30157869

RESUMO

BACKGROUND: In Parkinson's disease (PD), loss of striatal dopaminergic (DA) terminals and degeneration of DA neurons in the substantia nigra (SN) are associated with inflammation. Nucleotide-binding oligomerization domain-containing protein (NOD)2, one of the first discovered NOD-like receptors, plays an important role in inflammation. However, the role of NOD2 has not been elucidated in PD. METHODS: NOD2 mRNA and protein expression in the SN and the striatum of C57BL/6 mice treated with 6-hydroxydopamine (6-OHDA) was measured. We next investigated the potential contribution of the NOD2-dependent pathway to 6-OHDA-induced DA degeneration using NOD2-deficient (NOD2-/-) mice. Assays examining DA degeneration and inflammation include HPLC, Western blot, immunohistochemistry, TUNEL staining, and cytometric bead array. To further explore a possible link between NADPH oxidase 2 (NOX2) and NOD2 signaling in PD, microglia were transfected with shRNA specific to NOX2 in vitro and apocynin were given to mice subjected to 6-OHDA and muramyl dipeptide (MDP) striatal injection. RESULTS: The expression of NOD2 was upregulated in an experimental PD model induced by the neurotoxin 6-OHDA. NOD2 deficiency resulted in a protective effect against 6-OHDA-induced DA degeneration and neuronal death, which was associated with the attenuated inflammatory response. Moreover, silencing of NOX2 in microglia suppressed the expression of NOD2 and the inflammatory response induced by 6-OHDA and attenuated the toxicity of conditioned medium from 6-OHDA or MDP-stimulated microglia to neuronal cells. Furthermore, apocynin treatment inhibited NOD2 upregulation and DA degeneration in the SN of WT mice induced by 6-OHDA and MDP. CONCLUSION: This study provides the direct evidence that NOD2 is related to 6-OHDA-induced DA degeneration through NOX2-mediated oxidative stress, indicating NOD2 is a novel innate immune signaling molecule participating in PD inflammatory response.


Assuntos
Dopamina/metabolismo , Regulação da Expressão Gênica/genética , NADPH Oxidase 2/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Oxidopamina , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/complicações , Acetofenonas/farmacologia , Acetofenonas/uso terapêutico , Animais , Apomorfina/farmacologia , Linhagem Celular Transformada , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Degeneração Neural/etiologia , Degeneração Neural/fisiopatologia , Proteína Adaptadora de Sinalização NOD2/genética , Doença de Parkinson Secundária/genética , Doença de Parkinson Secundária/metabolismo , Espécies Reativas de Oxigênio/metabolismo
7.
Exp Neurol ; 302: 205-213, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29407460

RESUMO

Triggering receptor expressed on myeloid cells-2 (TREM2) was a newly identified receptor expressed on microglia. Several observations support the hypothesis that TREM2 variation may confer susceptibility to Parkinson's disease (PD). Therefore, in this paper, we explored the role of TREM2 in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Our results revealed that overexpression of TREM2 remarkably reduced MPTP-induced neuropathology including the dopaminergic neurodegeneration and neuroinflammation in vivo. Further mechanistic study revealed that TREM2 inhibited neuroinflammation by negatively regulating the TRAF6/TLR4-mediated activation of the MAPK and NF-κB signaling pathways. Taken together, our data suggest that TREM2 may have important neuroprotective effects against PD by critically modulating neuroinflammatory responses. These findings provide insights into the role of TREM2 in PD pathogenesis, and highlight TREM2 as a potential therapeutic target for this kind of disease.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Inflamação/metabolismo , Glicoproteínas de Membrana/metabolismo , Transtornos Parkinsonianos/patologia , Receptores Imunológicos/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Adenoviridae/genética , Animais , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Inflamação/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Transtornos Parkinsonianos/complicações , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
8.
J Cell Mol Med ; 22(2): 1103-1117, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28994231

RESUMO

Epithelial-mesenchymal transition (EMT) plays an important role in the invasiveness and metastasis of gastric cancer. Therefore, identifying key molecules involved in EMT will provide new therapeutic strategy for treating patients with gastric cancer. TIPE1 is a newly identified member of the TIPE (TNFAIP8) family, and its contributions to progression and metastasis have not been evaluated. In this study, we found that the levels of TIPE1 were significantly reduced and inversely correlated with differentiation status and distant metastasis in primary gastric cancer tissues. We further observed overexpression of TIPE1 in aggressive gastric cancer cell lines decreased their metastatic properties both in vitro and in vivo as demonstrated by markedly inhibiting EMT and metastasis of gastric cancer cells in nude mice. Consistently, gene silencing of TIPE1 in well-differentiated gastric cancer cell line (AGS) inhibited these processes. Mechanistically, we found that TIPE1-medicated Wnt/ß-catenin signalling was one of the critical signal transduction pathways that link TIPE1 to EMT inhibition. Importantly, TIPE1 dramatically restrained the expression and activities of MMP2 and MMP9 which are demonstrated to promote tumour progression and are implicated in EMT. Collectively, these findings provide new evidence for a better understanding of the biological activities of TIPE1 in progression and metastasis of gastric cancer and suggest that TIPE1 may be an innovative diagnostic and therapeutic target of gastric cancer.


Assuntos
Movimento Celular , Regulação para Baixo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Via de Sinalização Wnt , Animais , Carcinogênese/genética , Carcinogênese/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Nus , Pessoa de Meia-Idade , Modelos Biológicos , Invasividade Neoplásica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Neoplasias Gástricas/genética
9.
Nat Commun ; 8(1): 413, 2017 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-28871079

RESUMO

Podocyte injury is a major determinant of proteinuric kidney disease and the identification of potential therapeutic targets for preventing podocyte injury has clinical importance. Here, we show that histone deacetylase Sirt6 protects against podocyte injury through epigenetic regulation of Notch signaling. Sirt6 is downregulated in renal biopsies from patients with podocytopathies and its expression correlates with glomerular filtration rate. Podocyte-specific deletion of Sirt6 exacerbates podocyte injury and proteinuria in two independent mouse models, diabetic nephropathy, and adriamycin-induced nephropathy. Sirt6 has pleiotropic protective actions in podocytes, including anti-inflammatory and anti-apoptotic effects, is involved in actin cytoskeleton maintenance and promotes autophagy. Sirt6 also reduces urokinase plasminogen activator receptor expression, which is a key factor for podocyte foot process effacement and proteinuria. Mechanistically, Sirt6 inhibits Notch1 and Notch4 transcription by deacetylating histone H3K9. We propose Sirt6 as a potential therapeutic target for the treatment of proteinuric kidney disease.Podocytes are essential components of the renal glomerular filtration barrier and podocyte dysfunction leads to proteinuric kidney disease. Here Liu et al. show that Sirt6 protects podocytes from apoptosis and inflammation by increasing autophagic flux through inhibition of the Notch pathway.


Assuntos
Podócitos/metabolismo , Podócitos/patologia , Proteinúria/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Sirtuínas/deficiência , Acetilação , Animais , Apoptose , Autofagia , Biópsia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/patologia , Regulação para Baixo , Doxorrubicina , Deleção de Genes , Histonas/metabolismo , Humanos , Inflamação/patologia , Integrases/metabolismo , Rim/patologia , Lisina/metabolismo , Masculino , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Ratos , Sirtuínas/metabolismo
10.
Neurosci Lett ; 658: 114-120, 2017 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-28823893

RESUMO

OBJECTIVE: Histone deacetylase 6 (HDAC6) has been regarded as an unusual HDAC because of its unique properties. It contains two deacetylase catalytic domains and one ubiquitin-binding domain, thus exerting both enzymatic and non-enzymatic actions on cellular function. To date, the ubiquitin-binding activity of HDAC6 has been implicated in several neurodegenerative disorders including Parkinson's disease (PD). However, the deacetylation effect of HDAC6 in PD has not been fully illustrated. Therefore, the aim of the present study was to explore the role of deacetyation activity of HDAC6 in PD. METHODS: We used an in vivo 6-OHDA induced PD model and a specific HDAC6 inhibitor tubastatin A to investigate the acetylation levels of peroxiredoxin1 (Prx1) and peroxiredoxin2 (Prx2) and to explore the effects of tubastain A on nigrostriatal dopaminergic system. RESULTS: Our results showed that expression of HDAC6 significantly increased in dopaminergic neurons after 6-OHDA injury. Acetylation levels of Prx1 and Prx2 decreased. Pharmacological inhibition of HDAC6 with specific inhibitor tubastatin A increased acetylation of Prx1 and Prx2, reduced ROS production and ameliorated dopaminergic neurotoxicity. CONCLUSION: Our results for the first time provide evidence that HDAC6 medicated deacetylation of Prx1 and Prx2 contributes to oxidative injury in PD, suggesting that the development of specific HDAC6 inhibitor is required to develop more effective therapeutic strategies to treat PD.


Assuntos
Acetilação/efeitos dos fármacos , Desacetilase 6 de Histona/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Peroxirredoxinas/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Lesões Encefálicas/metabolismo , Dopamina/metabolismo , Ácidos Hidroxâmicos/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo
11.
Eur J Pharmacol ; 815: 98-108, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28844872

RESUMO

Losartan, an angiotensin (Ang) II type 1 receptor blocker (ARB), has been revealed to protect against cerebral ischemia/reperfusion (I/R) injury. However, the mechanism by which losartan protect brain ischemia injury is still obscure. In this study, we investigated whether losartan protected against cerebral I/R injury by reducing apoptosis and the possible signaling pathways. Wistar rats were pretreated for 14 days with 5mg/kg losartan, and then subjected to middle cerebral artery occlusion (MCAO) for 2h followed by reperfusion. Meanwhile, PC12 cells pretreated with losartan were exposed to oxygen-glucose deprivation-reoxygenation (OGD/R), an in vitro model of cerebral ischemia. Our results showed that administration of losartan significantly inhibited the apoptosis by decreasing the number of apoptotic cells, decreasing the protein level of cleaved caspase-3, cytochrom C and Bax, and increasing the level of Bcl-2 both in vivo and in vitro. Moreover, losartan treatment markedly enhanced the phosphorylation of Akt and blockade of PI3K activity by wortmannin dramatically inhibited Akt phosphorylation and attenuated the anti-apoptotic effect of losartan. Furthermore, pretreatment with losartan significantly increased the protein level of ß-arrestin1 and silence of ß-arrestin1 by siRNA partly attenuated losartan-induced anti-apoptotic effect and the phosphorylation of Akt. These results suggested that ß-arrestin1 modulated the activation of Akt in losartan-induced anti-apoptotic effect in cerebral I/R. Our data would provide a new molecular basis for further understanding of protective effect of losartan in cerebral I/R injury and may provide benefits of using losartan in the treatment of cerebrovascular disease.


Assuntos
Apoptose/efeitos dos fármacos , Isquemia Encefálica/complicações , Losartan/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/tratamento farmacológico , beta-Arrestina 1/metabolismo , Animais , Citoproteção/efeitos dos fármacos , Losartan/uso terapêutico , Masculino , Células PC12 , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia
12.
Oxid Med Cell Longev ; 2017: 7191856, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29387294

RESUMO

Although studies have seen dramatic advances in the understanding of the pathogenesis of stroke such as oxidative stress, inflammation, excitotoxicity, calcium overload and apoptosis, the delivery of stroke therapies is still a great challenge. In this study, we designed and synthesized a series of novel twin compounds containing tetramethylpyrazine and carnitine substructures and explored their therapeutic potential and mechanism in stroke-related neuronal injury. We first screened the neuroprotective effects of candidate compounds and found that among the tested compounds, LR134 and LR143 exhibited significant neuroprotection as evidenced by reducing cerebral infarct and edema, improving neurological function as well as blood-brain barrier integrity in rats after cerebral ischemia/reperfusion injury. We further demonstrated that the neuroprotective effects of compounds LR134 and LR143 were associated with the reduced inflammatory responses and NADPH oxidase- (NOX2-) mediated oxidative stress and the protection of mitochondria accompanied by the improvement of energy supply. In summary, this study provides direct evidence showing that the novel twin compounds containing tetramethylpyrazine and carnitine substructures have neuroprotective effects with multiple therapeutic targets, suggesting that modulation of these chemical structures may be an innovative therapeutic strategy for treating patients with stroke.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Carnitina/farmacologia , Fármacos Neuroprotetores/farmacologia , Pirazinas/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Carnitina/química , Modelos Animais de Doenças , Masculino , NADPH Oxidase 2/metabolismo , Fármacos Neuroprotetores/química , Pirazinas/química , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia
13.
Neurochem Res ; 42(2): 686-696, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27943027

RESUMO

FTY720 (fingolimod) is the first oral drug approved for treating relapsing-remitting forms of multiple sclerosis. It is also protective in other neurological models including ischemia, Alzheimer's disease, Huntington disease and Rett syndrome. However, whether it might protect in a 6-hydroxydopamine (6-OHDA) mouse model associated with the dopaminergic pathology of Parkinson's disease (PD), has not been explored. Therefore, in the present study, we investigated the effects of FTY720 on 6-OHDA-induced neurotoxicity in cell cultures and mice. Here we show that FTY720 protected against 6-OHDA cytotoxicity and apoptosis in SH-SY5Y cells. We also show that prior administration of FTY720 to 6-OHDA lesioned mice ameliorated both motor deficits and nigral dopaminergic neurotoxicity, while also reducing 6-OHDA-associated inflammation. The protective effects of FTY720 were associated with activation of AKT and ERK1/2 pro-survival pathways and an increase in brain derived neurotrophic factor (BDNF) expression in vitro and in vivo. These findings suggest that FTY720 holds promise as a PD therapeutic acting, at least in part, through AKT/ERK1/2/P-CREB-associated BDNF expression.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Cloridrato de Fingolimode/uso terapêutico , Degeneração Neural/metabolismo , Degeneração Neural/prevenção & controle , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/prevenção & controle , Animais , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Relação Dose-Resposta a Droga , Cloridrato de Fingolimode/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxidopamina/toxicidade , Transtornos Parkinsonianos/induzido quimicamente
14.
Neurochem Res ; 42(4): 1141-1150, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28005221

RESUMO

Parkinson's disease (PD) is a progressive neurodegenerative disorder and characterized by motor system disorders resulting in loss of dopaminergic (DA) neurons. CXC195, a novel tetramethylpyrazine derivative, has been shown strongest neuroprotective effects due to its anti-apoptotic activity. However, whether CXC195 protects against DA neuronal damage in PD and the mechanisms underlying its beneficial effects are unknown. The purpose of our study was to investigate the potential neuroprotective role of CXC195 and to elucidate its mechanism of action against 6-hydroxydopamine (6-OHDA)-induced mouse model of PD. CXC195 administration improved DA neurodegeneration in PD mice induced by 6-OHDA. Our further findings confirmed treatment of CXC195 at the dose of 10 mg/kg significantly inhibited the apoptosis by decreasing the level of cleaved caspase-3 and Bax, and increasing the level of Bcl-2 in 6-OHDA-lesioned mice. Meanwhile, 6-OHDA also decreased the amount of phosphorylated Akt while increased GSK-3ß activity (the amount of phosphorylated GSK-3ß at Ser9 was decreased) which was prevented by CXC195. Wortmannin, a specific PI3K inhibitor, dramatically abolished the changes induced by CXC195. Our study firstly demonstrated that CXC195 protected against DA neurodegeneration in 6-OHDA-induced PD model by its anti-apoptotic properties and PI3K/Akt/GSK3ß signaling pathway was involved in it.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Transtornos Parkinsonianos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Piperazinas/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirazinas/uso terapêutico , Animais , Apoptose , Neurônios Dopaminérgicos/efeitos dos fármacos , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Oxidopamina/toxicidade , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/tratamento farmacológico , Piperazinas/química , Piperazinas/farmacologia , Pirazinas/química , Pirazinas/farmacologia , Distribuição Aleatória , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
15.
Hypertension ; 69(2): 259-266, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27872232

RESUMO

Hyperhomocysteinemia (hHcys) is an important independent risk factor for the development of cardiovascular disease and end-stage renal disease. Although multiple approaches lowering the levels of homocysteine have been used in experimental studies and clinical trials, there is no effective therapy available to fully prevent homocysteine-induced injury. Therefore, identifying key molecules in the pathogenic pathways may provide clues to develop new therapeutic strategies for the treatment of hHcys-associated injury beyond lowering the plasma homocysteine levels. In this study, we found that the levels of progranulin (PGRN), an autocrine growth factor, were significantly reduced in the kidney and heart from a mouse model of hHcys. We further observed that in hHcys, PGRN-deficient mice significantly exacerbated cardiorenal injury as evidenced by higher levels of urinary albumin excretion, more severe renal morphological injuries, including pronounced glomerular basement membrane thickening and podocyte foot process effacement, and adverse myocardial remodeling versus wild-type mice. Mechanistically, we found that PGRN-medicated Wnt/ß-catenin signaling was one of the critical signal transduction pathways that links homocysteine to cardiorenal injury. Importantly, we finally provided direct evidence for the therapeutic potential of PGRN in mice with hHcys by pretreatment with recombinant human PGRN. Collectively, our results suggest that PGRN may be an innovative therapeutic strategy for treating patients with hHcys.


Assuntos
Síndrome Cardiorrenal/prevenção & controle , Hiper-Homocisteinemia/complicações , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Animais , Síndrome Cardiorrenal/diagnóstico , Síndrome Cardiorrenal/etiologia , Modelos Animais de Doenças , Ecocardiografia , Granulinas , Homocisteína/sangue , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/tratamento farmacológico , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Progranulinas , Proteínas Recombinantes/uso terapêutico
16.
Biochem Biophys Res Commun ; 479(4): 656-662, 2016 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-27693696

RESUMO

Nucleotide binding oligomerization domain (NOD)-like receptors (NLRs) are a class of cytoplasmic pattern-recognition receptors with a major role in innate immunity. Fourteen of the twenty-two human NLRs contain a pyrin domain and form the NLRP subfamily (NLRPs). Among NLRPs, NLRP2 is less well-understood in aspects of distribution and functions, especially in central nervous system (CNS). This study was the first to explore the expression of NLRP2 in central nervous system both under normal conditions and in ischemic stroke models. We found NLRP2 protein had a basal level of expression in CNS, mainly in astrocytes and was significantly elevated in ischemic brains in vivo or oxygen-glucose deprivation-treated cells in vitro. And silencing of NLRP2 genes could reduce the apoptotic rate of oxygen-glucose deprivation-treated cells. Thus high expression of NLRP2, especially in astrocytes, may play important roles in the pathophysiological process of ischemic stroke and has potential clinical value for the treatment of ischemic stroke.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Proteínas/metabolismo , Acidente Vascular Cerebral/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose , Astrócitos/patologia , Encéfalo/patologia , Modelos Animais de Doenças , Inativação Gênica , Glucose/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular , Proteínas/genética , RNA Interferente Pequeno/genética , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia , Regulação para Cima
17.
J Cell Mol Med ; 20(6): 1139-49, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26865248

RESUMO

Histone deacetylase (HDAC) 9, a member of class II HDACs, regulates a wide variety of normal and abnormal physiological functions, which is usually expressed at high levels in the brain and skeletal muscle. Although studies have highlighted the importance of HDAC-mediated epigenetic processes in the development of ischaemic stroke and very recent genome-wide association studies have identified a variant in HDAC9 associated with large-vessel ischemic stroke, the molecular events by which HDAC9 induces cerebral injury keep unclear. In this study, we found that HDAC9 was up-regulated in the ischaemic cerebral hemisphere after cerebral ischaemia/reperfusion (I/R) injury in rats and in vivo gene silencing of HDAC9 by recombinated lentivirus infection in the brain reduced cerebral injury in experimental stroke. We further demonstrated that HDAC9 contributed to oxygen-glucose deprivation-induced brain microvessel endothelial cell dysfunction as demonstrated by the increased inflammatory responses, cellular apoptosis and endothelial cell permeability dysfunction accompanied by reduced expression of tight-junction proteins. We further found that HDAC9 suppressed autophagy, which was associated with endothelial dysfunction. This study for the first time provides direct evidence that HDAC9 contributes to endothelial cell injury and demonstrates that HDAC9 is one of critical components of a signal transduction pathway that links cerebral injury to epigenetic modification in the brain.


Assuntos
Isquemia Encefálica/complicações , Isquemia Encefálica/enzimologia , Encéfalo/irrigação sanguínea , Células Endoteliais/patologia , Histona Desacetilases/metabolismo , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Animais , Apoptose , Autofagia , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/ultraestrutura , Isquemia Encefálica/patologia , Permeabilidade da Membrana Celular , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Inativação Gênica , Técnicas de Transferência de Genes , Glucose/deficiência , Histona Desacetilases/genética , Inflamação/patologia , Lentivirus/metabolismo , Masculino , Microvasos/patologia , Oxigênio , Ratos Sprague-Dawley , Traumatismo por Reperfusão/genética , Regulação para Cima
18.
Neurochem Res ; 41(4): 779-86, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26537816

RESUMO

In this study, we investigated whether tanshinone I (T-I) has therapeutic effects in cellular and animal model of Parkinson's disease (PD), and explore its possible mechanism. For this purpose, human neuroblastoma SH-SY5Y cells were cultured and exposed to 100 µM 6-hydroxydopamine (6-OHDA) in the absence or presence of T-I (1, 2.5 and 5 µM). The results revealed that 6-OHDA-induced cell death was reduced by T-I pretreatment as measured by MTT assay, lactate dehydrogenase release and flow cytomety analysis of cell apoptosis. The increase in the reactive oxygen species caused by 6-OHDA treatment was also attenuated by T-I in SH-SY5Y cells. T-I pretreatment was also shown to result in an increase in nuclear factor erythroid-2-related factor 2 (Nrf2) protein levels and its transcriptional activity as well as the upregulation of Nrf2-dependent genes encoding the antioxidant enzymes heme oxygenase-1, glutathione cysteine ligase regulatory subunit and glutathione cysteine ligase modulatory subunit in SH-SY5Y cells. Moreover, in the in vivo experiment, T-I treatment significantly attenuated 6-OHDA-induced striatal oxidative stress and ameliorated dopaminergic neurotoxicity in 6-OHDA-lesioned mice, as evidenced by western blot analysis of tyrosine hydroxylase (TH) and TH immunostaining of dopaminergic neurons in the substantia nigra and the striatum. Taken together, the results suggest that T-I may be beneficial for the treatment of neurodegenerative diseases like PD.


Assuntos
Abietanos/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Oxidopamina , Doença de Parkinson/prevenção & controle , Abietanos/uso terapêutico , Animais , Elementos de Resposta Antioxidante , Morte Celular , Linhagem Celular Tumoral , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/patologia , Fator 2 Relacionado a NF-E2/genética , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Transcrição Gênica , Regulação para Cima
19.
Fundam Clin Pharmacol ; 29(5): 478-87, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26182951

RESUMO

Ligustrazine is the most abundant and bioactive ingredient in Rhizoma Chuanxiong, a Chinese medicinal herb commonly used for the treatment of cardiovascular diseases. Chf197 is one of the structurally modified ligustrazine derivatives in a purpose of overcoming the rapid metabolism and short half-life of original. The plasma and urine pharmacokinetics of Chf197 in rats were studied after intravenous or intraperitoneal injection of Chf197 with the validated RP-HPLC method. The pharmacokinetic parameters of Chf197 injected intravenously 20 mg/kg were as follows: Cmax , 1.44 ± 0.4 mg/L; Tmax , 0.08 h; t1/2 , 3.03 ± 1.67 h; AUC, 3.85 ± 3.88 h/L; Vd , 31.66 ± 11.79L/kg; and CL, 9.29 ± 4.92 l/h/kg. Dose-dependent pharmacokinetics was observed, and a significantly higher dose-normalized AUC after intravenous administration was obtained than that after intraperitoneal administration. A possible metabolite was detected at about 3.1 min, and full-scan mass spectrum was adopted to predict its possible structure.


Assuntos
Medicamentos de Ervas Chinesas/farmacocinética , Pirazinas/farmacocinética , Animais , Área Sob a Curva , Biotransformação , Cromatografia de Fase Reversa , Medicamentos de Ervas Chinesas/administração & dosagem , Meia-Vida , Injeções Intraperitoneais , Injeções Intravenosas , Masculino , Taxa de Depuração Metabólica , Metacrilatos , Pirazinas/administração & dosagem , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização por Electrospray
20.
Neurochem Res ; 40(7): 1463-71, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25994859

RESUMO

Oxidative stress plays an important role in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease (AD). Eriodictyol, a flavonoid isolated from the Chinese herb Dracocephalum rupestre, has long been established as an antioxidant. The present study was designed to investigate the effect of eriodictyol on ß-amyloid 25-35 peptide (Aß25-35)-induced oxidative cell death in primary neurons and to explore the role of the nuclear factor erythroid-2-related factor 2/antioxidant response element (Nrf2/ARE) pathway in this process. For this purpose, primary cultures of cortical neurons were exposed to 15 µM Aß25-35 in the absence or presence of eriodictyol (20, 40 and 80 µM). The results revealed that Aß25-35-induced cytotoxicity and apoptotic characteristics such as activation of JNK/p38 apoptotic signaling pathway were effectively attenuated by eriodictyol pretreatment. Eriodictyol treatment also resulted in an increase in Nrf2 protein levels and subsequent activation of ARE pathway genes in primary cultured neurons. The protective effects of eriodictyol were attenuated by RNA interference-mediated knockdown of Nrf2 expression. Taken together, these results clearly demonstrate that eriodictyol protects neurons against Aß25-35-induced cell death partially through Nrf2/ARE signaling pathway, which further supports that eriodictyol might be a promising novel therapeutic agent for AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Morte Celular/efeitos dos fármacos , Flavanonas/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo , Fragmentos de Peptídeos/toxicidade , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Neurônios/metabolismo , Transporte Proteico , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...