Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cancer Cell ; 41(4): 693-710.e8, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-36963400

RESUMO

Malignant gliomas are largely refractory to immune checkpoint blockade (ICB) therapy. To explore the underlying immune regulators, we examine the microenvironment in glioma and find that tumor-infiltrating T cells are mainly confined to the perivascular cuffs and express high levels of CCR5, CXCR3, and programmed cell death protein 1 (PD-1). Combined analysis of T cell clustering with T cell receptor (TCR) clone expansion shows that potential tumor-killing T cells are mainly categorized into pre-exhausted/exhausted and effector CD8+ T subsets, as well as cytotoxic CD4+ T subsets. Notably, a distinct subpopulation of CD4+ T cells exhibits innate-like features with preferential interleukin-8 (IL-8) expression. With IL-8-humanized mouse strain, we demonstrate that IL-8-producing CD4+ T, myeloid, and tumor cells orchestrate myeloid-derived suppressor cell infiltration and angiogenesis, which results in enhanced tumor growth but reduced ICB efficacy. Antibody-mediated IL-8 blockade or the inhibition of its receptor, CXCR1/2, unleashes anti-PD-1-mediated antitumor immunity. Our findings thus highlight IL-8 as a combinational immunotherapy target for glioma.


Assuntos
Glioma , Inibidores de Checkpoint Imunológico , Interleucina-8 , Animais , Camundongos , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Glioma/tratamento farmacológico , Glioma/patologia , Inibidores de Checkpoint Imunológico/farmacologia , Imunoterapia/métodos , Interleucina-8/metabolismo , Linfócitos T , Microambiente Tumoral
2.
Cell Mol Neurobiol ; 42(1): 265-278, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32653974

RESUMO

Long non-coding RNAs (lncRNAs) can not only regulate gene transcription and translation, but also participate in the development of central nervous system diseases as epigenetic modification factors. However, their functional significance in atherosclerosis-induced ischemic stroke (AIIS) is unclear. The study aimed to screen out differentially expressed lncRNAs (delncRNAs), and to elucidate their potential regulatory mechanisms in the pathophysiology of AIIS. Based on the clinicopathological features and clinical images, we screened out 10 patients with AIIS and recruited 10 healthy volunteers. Then we used microarray to detect the whole blood RNA of subjects, and explored the biological functions of delncRNAs by GO and KEGG analysis. After further analyzing the delncRNAs of THP-1 stimulated with ox-LDL, selective lncRNAs were screened and a corresponding lncRNA-mRNA interaction network was constructed through co-expression analysis. We yielded 180 delncRNAs (44 up-regulated and 136 down-regulated) and 218 demRNAs (45 up-regulated and 173 down-regulated). Lnc-SCARNA8 and lnc-SNRPN-2 are the most significant elevated and decreased lncRNA in AIIS, respectively. The delncRNAs may play a significant role in ubiquitination-mediated protein degradation signaling pathways. According to lncRNA-mRNA network, the expression of vacuolar protein sorting 13 homolog B (VPS13B) and biliverdin reductase B (BLVRB) were significantly regulated. Our findings suggest that the ubiquitinated proteasome pathway, VPS13B and BLVRB may play a fundamental role in the pathological process of AIIS.


Assuntos
Aterosclerose , AVC Isquêmico , RNA Longo não Codificante , Aterosclerose/complicações , Aterosclerose/genética , Redes Reguladoras de Genes , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transcriptoma/genética
3.
Pharmacol Res ; 169: 105640, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33915296

RESUMO

AIM: Brain microvascular endothelial cells (BMVECs), as the important structure of blood-brain barrier (BBB), play a vital role in ischemic stroke. Pyroptosis of different cells in the brain may aggravate cerebral ischemic injury, and PGC-1α plays a major role in pyroptosis. However, it is not known whether BMVECs undergo pyroptosis after ischemic stroke and whether PGC-1α activator Medioresinol (MDN) we discovered may be useful against pyroptosis of endothelial cells and ischemic brain injury. METHODS: For in vitro experiments, the bEnd.3 cells and BMVECs under oxygen and glucose-deprivation (OGD) were treated with or without MDN, and the LDH release, tight junction protein degradation, GSDMD-NT membrane location and pyroptosis-associated proteins were evaluated. For in vivo experiments, mice underwent transient middle cerebral artery occlusion (tMCAO) for ischemia model, and the neuroprotective effects of MDN were measured by infarct volume, the permeability of BBB and pyroptosis of BMVECs. For mechanistic study, effects of MDN on the accumulation of phenylalanine, mitochondrial reactive oxygen species (mtROS) were tested by untargeted metabolomics and MitoSOX Red probe, respectively. RESULTS: BMVECs underwent pyroptosis after ischemia. MDN dose-dependently activated PGC-1α, significantly reduced pyroptosis, mtROS and the expressions of pyroptosis-associated proteins (NLRP3, ASC, cleaved caspase-1, IL-1ß, GSDMD-NT), and increased ZO-1 and Occludin protein expressions in BMVECs. In tMCAO mice, MDN remarkably reduced brain infarct volume and the permeability of BBB, inhibited pyroptosis of BMVECs, and promoted long-term neurobehavioral functional recovery. Mechanistically, MDN promoted the interaction of PGC-1α with PPARα to increase PPARα nuclear translocation and transcription activity, further increased the expression of GOT1 and PAH, resulting in enhanced phenylalanine metabolism to reduce the ischemia-caused phenylalanine accumulation and mtROS and further ameliorate pyroptosis of BMVECs. CONCLUSION: In this study, we for the first time discovered that pyroptosis of BMVECs was involved in the pathogenesis of ischemic stroke and MDN as a novel PGC-1α activator could ameliorate the pyroptosis of endothelial cells and ischemic brain injury, which might attribute to reduction of mtROS through PPARα/GOT1 axis in BMVECs. Taken together, targeting endothelial pyroptosis by MDN may provide alternative therapeutics for brain ischemic stroke.


Assuntos
Aspartato Aminotransferase Citoplasmática/metabolismo , Endotélio Vascular/efeitos dos fármacos , AVC Isquêmico/tratamento farmacológico , Lignanas/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , PPAR alfa/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/agonistas , Piroptose/efeitos dos fármacos , Animais , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Imunofluorescência , Cromatografia Gasosa-Espectrometria de Massas , Células HEK293/efeitos dos fármacos , Humanos , Lignanas/farmacologia , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Fármacos Neuroprotetores/farmacologia , Ratos Sprague-Dawley
4.
Acta Pharmacol Sin ; 41(5): 730, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32081978

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Nano Lett ; 20(3): 1542-1551, 2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-32039606

RESUMO

Neuroinflammation plays a pivotal part in the pathogenesis of stroke. Orphan nuclear receptor NR4A1 is involved in the inflammatory response of microglia and macrophages. In this study, we discovered an old drug, 9-aminoacridine (9-AA), as a novel NR4A1 activator from our in-house FDA-approved drug library, which exhibited anti-inflammatory activities through an NR4A1/IL-10/SOCS3 signaling pathway and modulated the microglia activation. To improve the druggability of 9-AA, different liposomal formulations were screened and investigated. 9-AA-loaded liposome (9-AA/L) was prepared to reduce the adverse effect of 9-AA. Furthermore, 9-AA-loaded PEG/cRGD dual-modified liposome (9-AA/L-PEG-cRGD) was obtained, which displayed prolonged circulation, improved biodistribution, and increased brain accumulation. In the transient middle cerebral artery occlusion (tMCAO) rat model, 9-AA/L-PEG-cRGD significantly reduced brain infarct area, ameliorated ischemic brain injury, and promoted long-term neurological function recovery. This "from drug discovery to drug delivery" methodology provides a potential therapeutic strategy using the liposomal 9-AA, the NR4A1 activator to suppress neuroinflammation for treatment of ischemic stroke.


Assuntos
Aminacrina , Descoberta de Drogas , AVC Isquêmico/tratamento farmacológico , Aminacrina/química , Aminacrina/farmacocinética , Aminacrina/farmacologia , Animais , Células HEK293 , Humanos , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Lipossomos , Masculino , Camundongos , Microglia/metabolismo , Microglia/patologia , Proteínas do Tecido Nervoso/metabolismo , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacocinética , Peptídeos Cíclicos/farmacologia , Polietilenoglicóis/química , Polietilenoglicóis/farmacocinética , Polietilenoglicóis/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
7.
Cell Mol Neurobiol ; 39(2): 265-286, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30607811

RESUMO

Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long non-coding RNA contributing to protect the blood-brain barrier (BBB) after stroke. We searched for small molecules that may up-regulate MALAT1 and focused on polydatin (PD), a natural product, as a possible candidate. PD enhanced MALAT1 gene expression in rat brain microvascular endothelial cells, reducing cell toxicity and apoptosis after oxygen and glucose deprivation (OGD). These effects correlated with reduction of inflammatory factors and enhancement of expression of BBB markers. We found opposite changes after MALAT1 silencing. We determined that C/EBPß is a key transcription factor for PD-mediated MALAT1 expression. PPARγ activity is involved in MALAT1 protective effects through its coactivator PGC-1α and the transcription factor CREB. This suggests that PD activates the MALAT1/CREB/PGC-1α/PPARγ signaling pathway to protect endothelial cells against ischemia. PD administration to rats subjected to brain ischemia by transient middle cerebral artery occlusion (tMCAO) reduced cerebral infarct volume and brain inflammation, protected cerebrovascular endothelial cells and BBB integrity. These effects correlated with increased expression of MALAT1, C/EBPß, and PGC-1α. Our results strongly suggest that the beneficial effects of PD involve the C/EBPß/MALAT1/CREB/PGC-1α/PPARγ pathway, which may provide a novel therapeutic strategy for brain ischemic stroke.


Assuntos
Encéfalo/irrigação sanguínea , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Glucosídeos/uso terapêutico , Microvasos/metabolismo , PPAR gama/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , RNA Longo não Codificante/genética , Estilbenos/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Sequência de Bases , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Glucosídeos/química , Glucosídeos/farmacologia , Células HEK293 , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Modelos Biológicos , RNA Longo não Codificante/metabolismo , Ratos Sprague-Dawley , Estilbenos/química , Estilbenos/farmacologia , Fatores de Tempo
8.
Biochem Pharmacol ; 156: 385-395, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30195731

RESUMO

Macrophages, which have various phenotypes and diverse functions, are becoming the target cells in inflammatory diseases. In this study, we evaluated the effects of the natural product smiglaside A, a phenylpropanoid glycoside isolated from the traditional Chinese medicinal herb Smilax riparia, on macrophage polarization and investigated the underlying mechanisms. We found that smiglaside A promoted M2 polarization and reduced M1 polarization in LPS-stimulated RAW264.7 cells and primary mouse peritoneal macrophages. Further mechanistic studies showed that the promoting effect of smiglaside A on M2 polarization was attenuated by pharmacological inhibition or gene silencing of AMP-activated protein kinase (AMPK) or peroxisome proliferator-activated receptor γ (PPARγ). Moreover, smiglaside A-enhanced PPARγ activity was prevented by the AMPK inhibitor compound C and by an siRNA. These findings indicate that the AMPK-PPARγ pathway is involved in promotion of M2 macrophages by smiglaside A. In a mouse model of LPS-induced acute lung injury, smiglaside A significantly increased the survival rate of LPS-injected mice and ameliorated the LPS-induced inflammatory response and lung damage. In addition, smiglaside A enhanced the protein expression levels of phosphorylated AMPK and PPARγ in the lung and promoted alveolar macrophages to the M2 phenotype in this mouse model. Taken together, our results indicate that smiglaside A can promote macrophage polarization to an anti-inflammatory M2 phenotype via stimulating the AMPK-PPARγ signaling pathway. Our study may provide novel approaches and/or targets for drug development to treat inflammatory diseases such as acute lung injury and sepsis.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Furanos/farmacologia , Glicosídeos/farmacologia , Lipopolissacarídeos/toxicidade , Macrófagos Alveolares/efeitos dos fármacos , PPAR gama/metabolismo , Proteínas Quinases/metabolismo , Piranos/farmacologia , Quinases Proteína-Quinases Ativadas por AMP , Lesão Pulmonar Aguda/tratamento farmacológico , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Glicosídeos/química , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , PPAR gama/genética , Proteínas Quinases/genética , Células RAW 264.7 , Smilax/química
9.
Gene ; 649: 32-39, 2018 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-29391273

RESUMO

Long noncoding RNAs is a novel class of RNA molecules, which is closely related to the occurrence and development of human disease. Recent studies have highlighted the importance of MEG3 in angiogenesis and the maintenance of normal function of vascular endothelial cells. However, whether MEG3 contributes to human endothelial cell angiogenesis as well as potential mechanisms are largely unknown. In this work, we found that the high expression level of MEG3 in primary HUVEC was controlled by DNA methylation, and its expression in HUVEC was regulated by HIF-1α under hypoxia condition. Meanwhile, we discovered that knockdown of MEG3 significantly suppressed VEGFR2 mRNA level, but had no influence on gene expression of VEGFR1, Notch1, DLL4 and Hes1. MEG3 reduction also suppressed VEGF-induced endothelial migration and angiogenesis. Furthermore, MEG3 knockdown reduced the tube formation and the spheroid sprouting of primary HUVEC under normoxic and hypoxic conditions. Altogether, MEG3 regulated by HIF-1α is required to maintain VEGFR2 expression in endothelial cells and plays a vital role for VEGFA-mediated endothelial angiogenesis.


Assuntos
Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Hipóxia Celular/fisiologia , Linhagem Celular , Metilação de DNA , Técnicas de Silenciamento de Genes , Células HEK293 , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células MCF-7 , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
Brain Behav Immun ; 67: 101-117, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28803158

RESUMO

Neuroinflammation plays a vital role in the pathological process of cerebral ischemic stroke, but currently there is no effective treatment. After ischemia, microglia-produced proinflammatory mediator expression contributes to the aggravation of neuroinflammation, while anti-inflammatory activation of microglia develops an anti-neuroinflammatory effect via secretion of anti-inflammatory factor. Promoting the anti-inflammatory activation of microglia might be an effective treatment of stroke. Previously, we discovered one derivative of the natural product (+)-balasubramide, compound 3C, that exhibits a remarkably anti-neuroinflammatory effect in vitro with unknown mechanisms. Thus in this study, we aimed to clarify its molecular mechanisms and determine whether compound 3C has a neuroprotective effect after ischemia via regulation on microglial inflammation. We found that compound 3C promoted the anti-inflammatory mediator expression and reduced the proinflammatory mediator expression in LPS-stimulated BV2 cells and mouse primary microglia cells, which were reversed by AMP-activated protein kinase (AMPK) inhibition or AMPK upstream calmodulin-dependent protein kinase kinase beta (CaMKKß) inhibition. Compound 3C also prevented LPS-stimulated JNK activation and enhanced PGC-1α activation in microglia, which was attenuated by AMPK inhibition. Additionally, compound 3C ameliorated depressive behaviors in LPS-induced neuroinflammatory mice by promoting the anti-inflammatory activation of microglia. Furthermore, we found that compound 3C markedly reduced brain infarct volume, improved the neurological deficit in rats with ischemia and reduced the activated microglia/macrophage cells in the ischemic area, which concomitantly enhanced the anti-inflammatory mediator expression. A mechanistic study showed that the compound 3C-mediated activation of CaMKKß, AMPK and PGC-1α is involved in the anti-neuroinflammatory and neuroprotective effects of 3C in the brain of LPS-treated mice and ischemic rats. Taken together, our results show that compound 3C could suppress neuroinflammation in vitro and in vivo by modulating microglial activation state through the CaMKKß-dependent AMPK/PGC-1α signaling pathway, and maybe further be developed as a promising new drug candidate for the treatment of brain disorders such as stroke associated with brain inflammation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Encefalite/metabolismo , Compostos Heterocíclicos com 3 Anéis/química , Microglia/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular , Depressão , Encefalite/prevenção & controle , Infarto da Artéria Cerebral Média/metabolismo , Mediadores da Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Ratos Sprague-Dawley , Transdução de Sinais
11.
Cell Mol Neurobiol ; 38(4): 929-939, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29159732

RESUMO

Oxidative stress is a great challenge to neurons following cerebral ischemia. PGC-1α has been shown to act as a potent modulator of oxidative metabolism. In this study, the effects of ZLN005, a small molecule that activate PGC-1α, against oxygen-glucose deprivation (OGD)- or ischemia-induced neuronal injury in vitro and in vivo were investigated. Transient middle cerebral artery occlusion (tMCAO) was performed in rats and ZLN005 was administered intravenously at 2 h, 4 h, or 6 h after ischemia onset. Infarct volume and neurological deficit score were detected to evaluate the neuroprotective effects of ZLN005. Well-differentiated PC12 cells, which were subjected to OGD for 2 h followed by reoxygenation for 22 h, were used as an in vitro ischemic model. Changes in expression of PGC-1α, its related genes, and antioxidant genes were determined by real-time quantitative PCR. The results showed that ZLN005 reduced cerebral infarct volume and improved the neurological deficit in rat with tMCAO, and significantly protected OGD-induced neuronal injury in PC12 cells. Furthermore, ZLN005 enhanced expression of PGC-1α in PC12 cells and in the ipsilateral hemisphere of rats with tMCAO. Additionally, ZLN005 increased antioxidant genes, including SOD1 and HO-1, and significantly prevented the ischemia-induced decrease in SOD activity. Taking together, the PGC-1α activator ZLN005 exhibits neuroprotective effects under ischemic conditions and molecular mechanisms possibly involve activation of PGC-1α signaling pathway and cellular antioxidant systems.


Assuntos
Benzimidazóis/farmacologia , Isquemia Encefálica/tratamento farmacológico , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Isquemia Encefálica/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Isquemia/tratamento farmacológico , Isquemia/metabolismo , Masculino , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Células PC12 , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
12.
Antioxid Redox Signal ; 28(2): 141-163, 2018 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-28747068

RESUMO

AIMS: Microglia-mediated neuroinflammation plays an important role in focal ischemic stroke, a disorder with no effective therapeutic agents. Since microglial polarization to the M2 phenotype and reduction of oxidative stress are mediated through AMP-activated protein kinase (AMPK) and nuclear factor erythroid 2-related factor 2 (Nrf2) activation, we assessed the dual therapeutic effect of AMPK and Nrf2 activation by a novel neuroprotectant HP-1c in the treatment of ischemic stroke. RESULTS: We developed a novel class of hybrids (HP-1a-HP-1f) of telmisartan and 2-(1-hydroxypentyl)-benzoate (HPBA) as a ring-opening derivative of NBP. The most promising hybrid, HP-1c, exhibited more potent anti-inflammatory and neuroprotective effects in vitro and reduced brain infarct volume and improved neurological deficits in a rat model of transient focal cerebral ischemia when compared with telmisartan alone, NBP alone, or a combination of telmisartan and NBP. HP-1c had a therapeutic window of up to 24 h, ameliorated ischemic cerebral injury in permanent focal cerebral ischemia, and improved motor function. The beneficial effects of HP-1c in ischemic stroke were associated with microglial polarization to the M2 phenotype and reduced oxidative stress. HP-1c also shifted the M1/M2 polarization in a mouse neuroinflammatory model. The anti-inflammatory and anti-oxidative effects of HP-1c were associated with AMPK-Nrf2 pathway activation for neuroprotection. We showed that HP-1c penetrates the brain, has a plasma half-life of around 3.93 h, and has no toxicity in mice. Innovation and Conclusion: Our study results suggest that HP-1c, with dual AMPK- and Nrf2-activating properties, may have potential in further studies as a novel therapy for ischemic stroke. Antioxid. Redox Signal. 28, 141-163.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Encefalite/etiologia , Encefalite/metabolismo , Microglia/metabolismo , Fator 2 Relacionado a NF-E2/agonistas , Fator 2 Relacionado a NF-E2/metabolismo , Acidente Vascular Cerebral/complicações , Animais , Antioxidantes , Isquemia Encefálica , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Infarto Cerebral , Modelos Animais de Doenças , Encefalite/imunologia , Feminino , Ativação de Macrófagos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Microglia/imunologia , Microglia/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , PPAR gama/metabolismo , Ratos , alfa-Defensinas/química , alfa-Defensinas/farmacologia
13.
Cell Mol Neurobiol ; 38(1): 53-71, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28534246

RESUMO

Microglia, of myeloid origin, play fundamental roles in the control of immune responses and the maintenance of central nervous system homeostasis. These cells, just like peripheral macrophages, may be activated into M1 pro-inflammatory or M2 anti-inflammatory phenotypes by appropriate stimuli. Microglia do not respond in isolation, but form part of complex networks of cells influencing each other. This review addresses the complex interaction of microglia with each cell type in the brain: neurons, astrocytes, cerebrovascular endothelial cells, and oligodendrocytes. We also highlight the participation of microglia in the maintenance of homeostasis in the brain, and their roles in the development and progression of age-related neurodegenerative disorders.


Assuntos
Encéfalo/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Animais , Encéfalo/patologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Homeostase/fisiologia , Humanos , Microglia/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/patologia
14.
Pharmazie ; 72(12): 707-713, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29441954

RESUMO

A series of novel 4,5-bisindolyl-1,2,4-triazol-3-ones were designed, prepared and evaluated for their glycogen synthase kinase (GSK)-3ß inhibitory activities. Compounds exhibited favorable inhibitory potency towards GSK-3ß kinase at the molecular level and in cells indicated by significantly reducing GSK-3ß substrate Tau phosphorylation at Ser396 in primary neurons showing the inhibition of cellular GSK-3ß. In an in vitro model of neuronal injury, compounds 6b, 6d and 6f prevented glutamate-induced neuronal death which was closely associated with cerebral ischemic stroke. Preliminary structure-activity relationship was examined and showed that different substituents on the indole ring had significant influences on the GSK-3ß inhibitory potency. These findings may provide new insights into the development of novel GSK-3ß inhibitors as neuroprotective agents.


Assuntos
Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Triazóis/farmacologia , Animais , Isquemia Encefálica/prevenção & controle , Simulação por Computador , Técnicas In Vitro , Neurônios/patologia , Fármacos Neuroprotetores/síntese química , Fármacos Neuroprotetores/química , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/prevenção & controle , Relação Estrutura-Atividade , Triazóis/síntese química , Triazóis/química
15.
Acta Pharmacol Sin ; 38(1): 29-40, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27840407

RESUMO

Recent studies focus on promoting neurite outgrowth to remodel the central nervous network after brain injury. Currently, however, there are few drugs treating brain diseases in the clinic by enhancing neurite outgrowth. In this study, we established an NGF-induced PC12 differentiation model to screen novel compounds that have the potential to induce neuronal differentiation, and further characterized 4,10-Aromadendranediol (ARDD) isolated from the dried twigs of the Baccharis gaudichaudiana plant, which exhibited the capability of promoting neurite outgrowth in neuronal cells in vitro. ARDD (1, 10 µmol/L) significantly enhanced neurite outgrowth in NGF-treated PC12 cells and N1E115 cells in a time-dependent manner. In cultured primary cortical neurons, ARDD (5, 10 µmol/L) not only significantly increased neurite outgrowth but also increased the number of neurites on the soma and the number of bifurcations. Further analyses showed that ARDD (10 µmol/L) significantly increased the phosphorylation of ERK1/2 and the downstream GSK-3ß, subsequently induced ß-catenin expression and up-regulated the gene expression of the Wnt ligands Fzd1 and Wnt3a in neuronal cells. The neurite outgrowth-promoting effect of ARDD in neuronal cells was abolished by pretreatment with the specific ERK1/2 inhibitor PD98059, but was partially reversed by XAV939, an inhibitor of the Wnt/ß-catenin pathway. ARDD also increased the expression of BDNF, CREB and GAP-43 in N1E115 cells, which was reversed by pretreatment with PD98059. In N1E115 cells subjected to oxygen and glucose deprivation (OGD), pretreatment with ARDD (1-10 µmol/L) significantly enhanced the phosphorylation of ERK1/2 and induced neurite outgrowth. These results demonstrated that the natural product ARDD exhibits neurite outgrowth-inducing activity in neurons via activation of the ERK signaling pathway, which may be beneficial to the treatment of brain diseases.


Assuntos
Proteína GAP-43/biossíntese , Proteína GAP-43/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Sesquiterpenos/farmacologia , Animais , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/biossíntese , Flavonoides/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Camundongos , Fator de Crescimento Neural/farmacologia , Neuritos/metabolismo , Neuritos/ultraestrutura , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Ratos , Sesquiterpenos/antagonistas & inibidores , Sesquiterpenos de Guaiano
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA