Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 566
Filtrar
1.
J Clin Neurosci ; 101: 239-243, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35640432

RESUMO

OBJECTIVE: To investigate the association between lipoprotein-associated phospholipase A2 (Lp-PLA2) concentration and the incidence of acute ischemic stroke (AIS) in patients with atrial fibrillation (AF). METHODS: A total of 257 patients admitted to the Kaifeng Central Hospital were enrolled in this study. Receiver operating characteristic (ROC) curve analysis and multivariate logistic regression analysis were used to determine the association between Lp-PLA2 and AIS in patients with AF. RESULTS: In AF group, plasma Lp-PLA2 concentrations were significantly higher in patients with AIS than in those without it (277.4 vs 155.1, p < 0.001). And in the group of AIS patients, patients with AF also had a significantly higher level of Lp-PLA2 concentration than those without (277.4 vs 204.2, p < 0.001). The analysis of the ROC curve showed a significant diagnostic value of Lp-PLA2 for the incidence of AIS in patients with AF (AUC = 0.840, 95% CI: 0.737-0.871, p < 0.001), and the optimal cut-off point was 220.5 ng/ml, with a sensitivity and specificity of 82.14% and 75.5%, respectively. All AF patients were divided into two subgroups: the high Lp-PLA2 group (≥220.5 ng/ml) and the low Lp-PLA2 group (<220.5 ng/ml). And multivariate logistic regression analysis showed that after adjustment of confounders, Lp-PLA2 (OR 12.48, 95%CI 5.73-27.16, p < 0.001) was independently associated with the incidence of AIS in patients with AF. CONCLUSIONS: Plasma Lp-PLA2 concentration was independently associated with the development of AIS in patients with AF. Lp-PLA2 is a potential biomarker for stratification of risk for AIS in patients with AF.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase , Fibrilação Atrial , AVC Isquêmico , Acidente Vascular Cerebral , 1-Alquil-2-acetilglicerofosfocolina Esterase/sangue , Fibrilação Atrial/sangue , Fibrilação Atrial/enzimologia , Biomarcadores/sangue , Humanos , AVC Isquêmico/sangue , AVC Isquêmico/enzimologia , Curva ROC , Fatores de Risco , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/enzimologia
2.
J Biol Chem ; 298(3): 101721, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35151685

RESUMO

Hypoxia-inducible factor prolyl 4-hydroxylases (HIF-P4Hs) regulate the hypoxic induction of >300 genes required for survival and adaptation under oxygen deprivation. Inhibition of HIF-P4H-2 has been shown to be protective in focal cerebral ischemia rodent models, while that of HIF-P4H-1 has no effects and inactivation of HIF-P4H-3 has adverse effects. A transmembrane prolyl 4-hydroxylase (P4H-TM) is highly expressed in the brain and contributes to the regulation of HIF, but the outcome of its inhibition on stroke is yet unknown. To study this, we subjected WT and P4htm-/- mice to permanent middle cerebral artery occlusion (pMCAO). Lack of P4H-TM had no effect on lesion size following pMCAO, but increased inflammatory microgliosis and neutrophil infiltration was observed in the P4htm-/- cortex. Furthermore, both the permeability of blood brain barrier and ultrastructure of cerebral tight junctions were compromised in P4htm-/- mice. At the molecular level, P4H-TM deficiency led to increased expression of proinflammatory genes and robust activation of protein kinases in the cortex, while expression of tight junction proteins and the neuroprotective growth factors erythropoietin and vascular endothelial growth factor was reduced. Our data provide the first evidence that P4H-TM inactivation has no protective effect on infarct size and increases inflammatory microgliosis and neutrophil infiltration in the cortex at early stage after pMCAO. When considering HIF-P4H inhibitors as potential therapeutics in stroke, the current data support that isoenzyme-selective inhibitors that do not target P4H-TM or HIF-P4H-3 would be preferred.


Assuntos
Barreira Hematoencefálica , Infarto da Artéria Cerebral Média , Doenças Neuroinflamatórias , Prolil Hidroxilases , Acidente Vascular Cerebral , Animais , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/metabolismo , Permeabilidade da Membrana Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/metabolismo , Camundongos , Doenças Neuroinflamatórias/enzimologia , Doenças Neuroinflamatórias/metabolismo , Permeabilidade , Prolil Hidroxilases/metabolismo , Inibidores de Prolil-Hidrolase/farmacologia , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
J Neurosci ; 42(1): 16-32, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-34764155

RESUMO

A fundamental regulator of neuronal network development and plasticity is the extracellular matrix (ECM) of the brain. The ECM provides a scaffold stabilizing synaptic circuits, while the proteolytic cleavage of its components and cell surface proteins are thought to have permissive roles in the regulation of plasticity. The enzymatic proteolysis is thought to be crucial for homeostasis between stability and reorganizational plasticity and facilitated largely by a family of proteinases named matrix metalloproteinases (MMPs). Here, we investigated whether MMP2 and MMP9 play a role in mediating adult primary visual cortex (V1) plasticity as well as stroke-induced impairments of visual cortex plasticity in mice. In healthy adult mice, selective inhibition of MMP2/9 for 7 d suppressed ocular dominance plasticity. In contrast, brief inhibition of MMP2/9 after a cortical stroke rescued compromised plasticity. Our data indicate that the proteolytic activity of MMP2 and MMP9 is critical and required to be within a narrow range to allow adult visual plasticity.SIGNIFICANCE STATEMENT Learning and recovery from injuries depend on the plasticity of neuronal connections. The brain's extracellular matrix (ECM) provides a scaffold for stabilizing synaptic circuits, while its enzymatic proteolysis is hypothesized to regulate homeostasis between stability and reorganizational plasticity. ECM digestion is facilitated by a family of matrix metalloproteinases (MMPs). Here, we show that treatments that inhibit MMP2/9 can either inhibit or rescue cortical plasticity depending on cortical state: in the visual cortex of healthy adult mice, inhibition of MMP2/9 suppressed cortical plasticity. In contrast, brief inhibition of MMP2/9 after a stroke rescued compromised plasticity. Our data provide strong evidence that an optimal level of MMP2/9 proteolytic activity is crucial for adult visual plasticity.


Assuntos
Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Plasticidade Neuronal/fisiologia , Córtex Visual Primário/enzimologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Acidente Vascular Cerebral/enzimologia
4.
Stroke ; 52(6): e250-e258, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33947213

RESUMO

Despite successful recanalization, a significant number of patients with ischemic stroke experience impaired local brain tissue reperfusion with adverse clinical outcome. The cause and mechanism of this multifactorial complication are yet to be understood. At the current moment, major attention is given to dysfunction in blood-brain barrier and capillary blood flow but contribution of exaggerated constriction of cerebral arterioles has also been suggested. In the brain, arterioles significantly contribute to vascular resistance and thus control of perfusion. Accordingly, pathological changes in arteriolar wall function can, therefore, limit sufficient reperfusion in ischemic stroke, but this has not yet received sufficient attention. Although an increased vascular tone after reperfusion has been demonstrated in several studies, the mechanism behind it remains to be characterized. Importantly, the majority of conventional mechanisms controlling vascular contraction failed to explain elevated cerebrovascular tone after reperfusion. We propose here that the Na,K-ATPase-dependent Src kinase activation are the key mechanisms responsible for elevation of cerebrovascular tone after reperfusion. The Na,K-ATPase, which is essential to control intracellular ion homeostasis, also executes numerous signaling functions. Under hypoxic conditions, the Na,K-ATPase is endocytosed from the membrane of vascular smooth muscle cells. This initiates the Src kinase signaling pathway that sensitizes the contractile machinery to intracellular Ca2+ resulting in hypercontractility of vascular smooth muscle cells and, thus, elevated cerebrovascular tone that can contribute to impaired reperfusion after stroke. This mechanism integrates with cerebral edema that was suggested to underlie impaired reperfusion and is further supported by several studies, which are discussed in this article. However, final demonstration of the molecular mechanism behind Src kinase-associated arteriolar hypercontractility in stroke remains to be done.


Assuntos
Reperfusão , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/terapia , Vasoconstrição/fisiologia , Quinases da Família src/metabolismo , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/enzimologia , Encéfalo/irrigação sanguínea , Encéfalo/enzimologia , Revascularização Cerebral/tendências , Humanos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Reperfusão/tendências , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/metabolismo , Vasoconstrição/efeitos dos fármacos , Quinases da Família src/antagonistas & inibidores
5.
Int J Mol Sci ; 22(6)2021 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-33805800

RESUMO

Cyclin-dependent kinases (CDKs) are involved in many crucial processes, such as cell cycle and transcription, as well as communication, metabolism, and apoptosis. The kinases are organized in a pathway to ensure that, during cell division, each cell accurately replicates its DNA, and ensure its segregation equally between the two daughter cells. Deregulation of any of the stages of the cell cycle or transcription leads to apoptosis but, if uncorrected, can result in a series of diseases, such as cancer, neurodegenerative diseases (Alzheimer's or Parkinson's disease), and stroke. This review presents the current state of knowledge about the characteristics of cyclin-dependent kinases as potential pharmacological targets.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Ciclo Celular/genética , Quinases Ciclina-Dependentes/genética , Neoplasias/tratamento farmacológico , Doença de Parkinson/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/genética , Cardiomegalia/tratamento farmacológico , Cardiomegalia/enzimologia , Cardiomegalia/genética , Cardiomegalia/patologia , Fármacos Cardiovasculares/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Regulação da Expressão Gênica , Humanos , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/enzimologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia
6.
Sci Rep ; 11(1): 4958, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33654156

RESUMO

Since the treatment window of thrombolytic therapy for stroke is limited, new therapy remains to be developed. We have recently developed low-intensity pulsed ultrasound (LIPUS) therapy to improve cognitive dysfunction in mouse models of vascular dementia and Alzheimer's disease. Here, we further aimed to examine whether our LIPUS therapy improves neurological recovery from ischemic stroke, and if so, to elucidate the mechanisms involved. In a mouse model of middle cerebral artery occlusion (MCAO), we applied LIPUS (32 cycles, 193 mW/cm2) to the whole brain 3 times in the first week (days 1, 3, and 5) after MCAO. We evaluated neurological functions using behavioral tests and performed histological analyses. Furthermore, to elucidate how LIPUS works within the injured brain, we also tested the effects of LIPUS in endothelial nitric oxide synthase (eNOS)-deficient (eNOS-/-) mice. In wild-type mice, the LIPUS therapy markedly improved neurological functions in the tightrope and rotarod tests at 28 days after MCAO. Histological analyses showed that the LIPUS therapy significantly increased the numbers of CD31-positive blood vessels in the perifocal lesion and doublecortin (DCX)-positive neurons in the ischemic striatum, indicating the angio-neurogenesis effects of the therapy. Importantly, these beneficial effects of the LIPUS therapy were totally absent in eNOS-/- mice. No adverse effects of the LIPUS therapy were noted. These results indicate that the LIPUS therapy improves neurological functions after stroke through enhanced neuro-angiogenesis in mice in vivo in an eNOS-dependent manner, suggesting that it could a novel and non-invasive therapeutic option for stroke.


Assuntos
Neovascularização Fisiológica , Neurogênese , Óxido Nítrico Sintase Tipo III , Acidente Vascular Cerebral , Terapia por Ultrassom , Ondas Ultrassônicas , Animais , Masculino , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo III/metabolismo , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/fisiopatologia , Acidente Vascular Cerebral/terapia
7.
J Stroke Cerebrovasc Dis ; 29(10): 105152, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32912518

RESUMO

Sirtuins, class III histone deacetylases, are involved in the regulation of tissue repair processes and brain functions after a stroke. The ability of some isoforms of sirtuins to circulate between the nucleus and cytoplasm may have various pathophysiological effects on the cells. In present work, we focused on the role of non-mitochondrial sirtuins SIRT1, SIRT2, and SIRT6 in the restoration of brain cells following ischemic stroke. Here, using a photothrombotic stroke (PTS) model in mice, we studied whether local stroke affects the level and intracellular localization of SIRT1, SIRT2, and SIRT6 in neurons and astrocytes of the intact cerebral cortex adjacent to the ischemic ipsilateral hemisphere and in the analogous region of the contralateral hemisphere at different time points during the recovery period after a stroke. We evaluated the co-localization of sirtuins with growth-associated protein-43 (GAP-43), the presynaptic marker synaptophysin (SYN) and acetylated α-tubulin (Ac-α-Tub), that are associated with brain plasticity and are known to be involved in brain repair after a stroke. The results show that during the recovery period, an increase in SIRT1 and SIRT2 levels occurred. The increase of SIRT1 level was associated with an increase in synaptic plasticity proteins, whereas the increase of SIRT2 level was associated with an acetylated of α-tubulin, that can reduce the mobility of neurites. SIRT6 co-localized with GAP-43, but not with SYN. Moreover, we showed that SIRT1, SIRT2, and SIRT6 are not involved in the PTS-induced apoptosis of penumbra cells. Taken together, our results suggest that sirtuins functions differ depending on cell type, intracellular localization, specificity of sirtuins isoforms to different substrates and nature of post-translational modifications of enzymes.


Assuntos
Astrócitos/enzimologia , Córtex Cerebral/enzimologia , Trombose Intracraniana/complicações , Plasticidade Neuronal , Neurônios/enzimologia , Sirtuína 1/metabolismo , Sirtuína 2/metabolismo , Sirtuínas/metabolismo , Acidente Vascular Cerebral/enzimologia , Animais , Apoptose , Astrócitos/patologia , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Masculino , Camundongos , Neurônios/patologia , Recuperação de Função Fisiológica , Transdução de Sinais , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Fatores de Tempo
8.
Brain Res Bull ; 162: 151-165, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32592806

RESUMO

Epigenetic processes play important roles in brain responses to ischemic injury. We studied effects of photothrombotic stroke (PTS, a model of ischemic stroke) on the intracellular level and cellular localization of histone deacetylases HDAC3, HDAC4 and HDAC6 in the rat brain cortex, and tested the potential neuroprotector ability of their inhibitors. The background level of HDAC3, HDAC4 and HDAC6 in the rat cerebral cortex was relatively low. HDAC3 localized in the nuclei of some neurons and few astrocytes. HDAC4 was found in the neuronal cytoplasm. After PTS, their levels in penumbra did not change, but HDAC4 appeared in the nuclei of some cells. Its level in the cytoplasmic, but not nuclear fraction of penumbra decreased at 24, but not 4 h after PTS. HDAC6 was upregulated in neurons and astrocytes in the PTS-induced penumbra, especially in the nuclear fraction. Unlike HDAC3 and HDAC4, HDAC6 co-localized with TUNEL-positive apoptotic cells. Inhibitory analysis confirmed the involvement of HDAC6, but not HDAC3 and HDAC4 in neurodegeneration. HDAC6 inhibitor HPOB, HDAC2/8 inhibitor α-phenyl tropolone, and non-specific histone deacetylase inhibitor sodium valproate, but not HDAC3 inhibitor BRD3308, or HDAC4 inhibitor LMK235, decreased PTS-induced infarction volume in the mouse brain, reduced apoptosis, and recovered the motor behavior. HPOB also restored PTS-impaired acetylation of α-tubulin. α-phenyl tropolone restored acetylation of histone H4 in penumbra cells. These results suggest that histone deacetylases HDAC6 and HDAC2 are the possible molecular targets for anti-ischemic therapy, and their inhibitors α-phenyl tropolone, HBOP and sodium valproate can be considered as promising neuroprotectors.


Assuntos
Córtex Cerebral/enzimologia , Desacetilase 6 de Histona/biossíntese , Inibidores de Histona Desacetilases/farmacologia , Trombose Intracraniana/enzimologia , Acidente Vascular Cerebral/enzimologia , Tropolona/farmacologia , Ácido Valproico/farmacologia , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Expressão Gênica , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases/biossíntese , Trombose Intracraniana/patologia , Trombose Intracraniana/prevenção & controle , Lasers Semicondutores/efeitos adversos , Masculino , Camundongos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ratos , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/prevenção & controle , Tropolona/uso terapêutico , Ácido Valproico/uso terapêutico
9.
J Neurochem ; 155(6): 679-696, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32415988

RESUMO

Environmental enrichment (EE) is a generally accepted strategy to promote stroke recovery and its beneficial effect is positively correlated with neuroplasticity. However, the mechanisms underlying it remain elusive. Histone deacetylase 2 (HDAC2), a negative regulator of neuroplasticity, is up-regulated after stroke. Thus, we hypothesized that HDAC2 may participate in EE-mediated stroke recovery. In this study, focal stroke was induced by photothrombosis in male mice exposing to EE or standard housing (SH) conditions. Recombinant virus vectors, including Ad-HDAC2-Flag, AAV-CAG-EGFP-Cre, LV-shHDAC2, or their controls were microinjected into the motor cortex at 3 days before stroke. Grid-walking and cylinder tasks were conducted to assess motor function. Western blot and immunostaining were used to uncover the mechanisms underlying EE-mediated stroke recovery. We found that EE exposure reversed stroke-induced HDAC2 up-regulation, implicating HDAC2 in EE-mediated functional recovery. Importantly, EE-dependent stroke recovery was counteracted by over-expressing HDAC2, and HDAC2 knockdown promoted functional recovery from stroke to the similar extent as EE exposure. Moreover, the knockdown of HDAC2 epigenetically enhanced expressions of neurotrophins and neuroplasticity-related proteins, with similar effects as EE, and consequently, whole brain and corticospinal tract (CST) rewiring. Together, our findings indicate that HDAC2 is critical for EE-dependent functional restoration. Precisely targeting HDAC2 may mimic EE and serve as a novel therapeutic strategy for stroke recovery.


Assuntos
Meio Ambiente , Histona Desacetilase 2/metabolismo , Recuperação de Função Fisiológica/fisiologia , Acidente Vascular Cerebral/enzimologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia
10.
Stroke ; 51(3): 975-985, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32078472

RESUMO

Background and Purpose- Microglia/macrophages (Mi/MΦ) can profoundly influence stroke outcomes by acquiring functionally dominant phenotypes (proinflammatory or anti-inflammatory; deleterious or salutary). Identification of the molecular mechanisms that dictate the functional status of Mi/MΦ after brain ischemia/reperfusion may reveal novel therapeutic targets for stroke. We hypothesized that activation of TAK1 (transforming growth factor beta-activated kinase 1), a key MAP3K upstream of multiple inflammation-regulating pathways, drives Mi/MΦ toward a proinflammatory phenotype and potentiates ischemia/reperfusion brain injury. Methods- Young adult mice were subjected to 1 hour of middle cerebral artery occlusion (MCAO) followed by reperfusion. TAK1 was targeted by tamoxifen-induced Mi/MΦ-specific knockout or administration of a selective inhibitor 5Z-7-Oxozeaenol after MCAO. Neurobehavioral deficits and long-term gray matter and white matter injury were assessed up to 35 days after MCAO. Mi/MΦ functional status and brain inflammatory profiles were assessed 3 days after MCAO by RNA-seq, flow cytometry, and immunohistochemistry. Results- TAK1 Mi/MΦ-specific knockout markedly ameliorated neurological deficits in the rotarod and cylinder tests for at least 35 days after MCAO. Mechanistically, RNA-seq of purified brain Mi/MΦ demonstrated that proinflammatory genes and their predicted biological functions were downregulated or inhibited in microglia and macrophages from TAK1 Mi/MΦ-specific knockout mice versus WT mice 3 days after MCAO. Consistent with the anti-inflammatory phenotype of Mi/MΦ-specific knockout, oxozeaenol treatment mitigated neuroinflammation 3 days after MCAO, manifested by less Iba1+/CD16+ proinflammatory Mi/MΦ and suppressed brain invasion of various peripheral immune cells. Oxozeaenol treatment beginning 2 hours after MCAO improved long-term sensorimotor and cognitive functions in the foot fault, rotarod, and water maze tests. Furthermore, Oxozeaenol promoted both gray matter and white matter integrity 35 days after MCAO. Conclusions- TAK1 promotes ischemia/reperfusion-induced inflammation, brain injury, and maladaptive behavior by enhancing proinflammatory and deleterious Mi/MΦ responses. Therefore, TAK1 inhibition is a promising therapy to improve long-term stroke outcomes.


Assuntos
Comportamento Animal , Lesões Encefálicas/enzimologia , Isquemia Encefálica/enzimologia , MAP Quinase Quinase Quinases/metabolismo , Traumatismo por Reperfusão/enzimologia , Acidente Vascular Cerebral/enzimologia , Animais , Lesões Encefálicas/genética , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , MAP Quinase Quinase Quinases/genética , Macrófagos , Camundongos , Camundongos Knockout , Microglia , Traumatismo por Reperfusão/genética , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia , Fatores de Tempo , Zearalenona/análogos & derivados , Zearalenona/farmacologia
11.
J Cardiovasc Pharmacol ; 75(4): 344-350, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31895872

RESUMO

OBJECTIVE: This study aimed to explore the association between genetic variations of CYP19A1 and stroke susceptibility in the Chinese Han population. METHODS: A total of 477 stroke patients and 480 healthy controls were recruited in this study. The genotyping of CYP19A1 polymorphisms (rs4646, rs6493487, rs1062033, rs17601876, and rs3751599) was performed by the Agena MassARRAY platform. Under logistic regression models, we evaluated the associations of CYP19A1 polymorphisms and stroke susceptibility by odds ratio and 95% confidence interval. RESULTS: Our study showed that rs4646 (codominant: P = 0.020; recessive: P = 0.016) and rs17601876 (allele: P = 0.044; codominant: P = 0.011; dominant: P = 0.009; recessive: P = 0.046) significantly decreased the risk of stroke. In the stratification analysis, rs4646 is associated with decreased stroke risk among the individuals older than 64 years (codominant: P = 0.028; recessive: P = 0.010) and women (codominant: P = 0.029; recessive: P = 0.029), whereas rs1062033 increased stroke risk in the subgroup of age 64 years and younger (recessive: P = 0.042). The rs17601876 polymorphism has a strong relationship with stroke susceptibility, which is age and gender dependent. In haplotype analysis, we found a block (rs17601876 and rs3751599), and Ars17601876Grs3751599 haplotype is related to an increased stroke risk (P < 0.05). In addition, CYP19A1 variations had effects on clinical characteristics. CONCLUSION: CYP19A1 polymorphisms were significantly associated with stroke susceptibility in the Chinese Han population.


Assuntos
Aromatase/genética , Polimorfismo de Nucleotídeo Único , Acidente Vascular Cerebral/genética , Idoso , Povo Asiático , Estudos de Casos e Controles , China/epidemiologia , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Medição de Risco , Fatores de Risco , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/etnologia
12.
Mol Neurobiol ; 57(1): 226-238, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31493239

RESUMO

In ischemic stroke, vascular occlusion rapidly induces tissue infarct. Over the ensuing hours, damage spreads to adjacent tissue and forms transition zone (penumbra), which is potentially salvageable. Epigenetic regulation of chromatin structure controls gene expression and protein synthesis. We studied the expression of histone deacetylases HDAC1 and HDAC2 in the penumbra at 4 or 24 h after photothrombotic stroke (PTS) in the rat brain cortex. PTS increased the expression of HDAC1 and HDAC2 in penumbra and caused the redistribution of HDAC1 but not HDAC2 from the neuronal nuclei to cytoplasm. In astrocytes, HDAC1 expression and localization did not change. In neurons, HDAC2 localized exclusively in nuclei, but in astrocytes, it was also observed in processes. PTS induced neuronal apoptosis in the penumbra. TUNEL-stained apoptotic neurons co-localized with HDAC2 but not HDAC1. These data suggest that HDAC2 may represent the potential target for anti-stroke therapy and its selective inhibition may be a promising strategy for the protection of the penumbra tissue after ischemic stroke.


Assuntos
Apoptose , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Luz , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/enzimologia , Trombose/complicações , Trombose/enzimologia , Animais , Astrócitos/metabolismo , Biomarcadores/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Ratos Wistar
13.
Neuromolecular Med ; 22(2): 194-209, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31802381

RESUMO

Stroke is the second largest cause of death worldwide. Angiotensin converting enzyme (ACE) gene has emerged as an important player in the pathogenesis of hypertension and consequently stroke. It encodes ACE enzyme that converts the inactive decapeptide angiotensin I to active octapeptide, angiotensin II (Ang II). Dysregulation in the expression of ACE gene, on account of genetic variants or regulation by miRNAs, alters the levels of ACE in the circulation. Variable expression of ACE affects the levels of Ang II. Ang II acts through different signal transduction pathways via various tyrosine kinases (receptor/non-receptor) and protein serine/threonine kinases, initiating a downstream cascade of molecular events. In turn these activated molecular pathways might lead to hypertension and inflammation thereby resulting in cardiovascular and cerebrovascular diseases including stroke. In order to regulate the overexpression of ACE, many ACE inhibitors and blockers have been developed, some of which are still under clinical trials.


Assuntos
Hipertensão/complicações , Peptidil Dipeptidase A/fisiologia , Acidente Vascular Cerebral/etiologia , Angiotensina II/fisiologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Bradicinina/metabolismo , Antagonistas dos Receptores da Bradicinina/farmacologia , Antagonistas dos Receptores da Bradicinina/uso terapêutico , Regulação Enzimológica da Expressão Gênica , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/genética , Macrófagos/fisiologia , Camundongos , MicroRNAs/genética , Mutação de Sentido Incorreto , Miócitos de Músculo Liso/fisiologia , Neuroglia/fisiologia , Peptidil Dipeptidase A/biossíntese , Peptidil Dipeptidase A/genética , Polimorfismo de Nucleotídeo Único , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/prevenção & controle
14.
Stroke ; 51(1): 224-231, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31726963

RESUMO

Background and Purpose- The role of dual-antiplatelet therapy with clopidogrel plus aspirin has been demonstrated to substantially decrease the risk of recurrent stroke among patients with minor stroke and transient ischemic attack. We aimed to determine whether the efficacy of clopidogrel-aspirin therapy among patients with minor stroke / transient ischemic attack was influenced by the stratification of CYP2C19 genotype and body mass index (BMI). Methods- CYP2C19 loss-of-function allele (LoFA) carriers were defined as patients with either LoFA of *2 or *3. Low/normal weight and overweight/obesity was defined as BMI <25 and ≥25 kg/m2, respectively. Primary outcome was defined as stroke recurrence at 3 months. Results- In a total of 2933 patients, there were 1726 (58.8%) LoFA carriers and 1275 (43.5%) patients with overweight/obesity (BMI ≥25 kg/m2). Stratified analyses by LoFA carrying status and BMI, hazard ratios (hazard ratios 95% CIs) of the clopidogrel-aspirin therapy for stroke recurrence were 0.90 (0.60-1.36), 0.87 (0.56-1.35), 0.65 (0.39-1.09), and 0.40 (0.22-0.71) among subgroups of LoFA carriers with overweight/obesity, LoFA carriers with low/normal weight, LoFA noncarriers with overweight/obesity, and LoFA noncarriers with low/normal weight, respectively, with P=0.049 for interaction. Conclusions- Efficacy of clopidogrel-aspirin therapy in reducing the risk of stroke recurrence is not present in CYP2C19 LoFA noncarriers with overweight/obesity. Our study suggests that BMI significantly influences the correlation between CYP2C19 genotype and efficacy of clopidogrel-aspirin therapy. Clinical Trial Registration- URL: https://www.clinicaltrials.gov. Unique identifier: NCT00979589.


Assuntos
Alelos , Aspirina/administração & dosagem , Clopidogrel/administração & dosagem , Citocromo P-450 CYP2C19/genética , Mutação com Perda de Função , Obesidade , Acidente Vascular Cerebral , Idoso , Índice de Massa Corporal , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade/tratamento farmacológico , Obesidade/enzimologia , Obesidade/genética , Fatores de Risco , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/genética
15.
J Thromb Thrombolysis ; 49(1): 10-17, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31643039

RESUMO

Prasugrel, a novel P2Y12 receptor antagonist, has been shown to be more effective than clopidogrel for preventing cardiovascular events in patients with acute coronary syndromes undergoing percutaneous coronary intervention. We investigated the dose-response antiplatelet effects of prasugrel compared with clopidogrel in Japanese patients with non-cardioembolic stroke. The influence of cytochrome P450 (CYP) polymorphisms on the antiplatelet effects of both drugs was also compared. In this multicenter randomized active-control comparative study, patients were randomized to receive prasugrel 2.5 mg, 5 mg, or 7.5 mg (double blind) or clopidogrel 75 mg (open label) once daily for 14 days. The primary endpoint was inhibition of platelet aggregation (IPA) in response to adenosine diphosphate 20 µM within 8 h of study drug administration on day 14. Of the 66 patients randomized, data from 63 (prasugrel 2.5 mg, 5 mg, and 7.5 mg groups, n = 14, 16, and 18, respectively; clopidogrel group, n = 15) were used in the pharmacodynamic assessment. IPA (arithmetic mean ± SD) after prasugrel administration increased dose-dependently (33 ± 9%, 44 ± 11%, and 53 ± 14%, at 2.5 mg, 5 mg, and 7.5 mg, respectively) and was higher in these groups than after clopidogrel (23 ± 16%). In a subgroup of CYP2C19 intermediate metabolizers, IPA was higher in the prasugrel 5 mg and 7.5 mg groups than in the clopidogrel group. No death or serious adverse events were reported. Prasugrel was well tolerated at doses up to 7.5 mg/day and had antiplatelet effects higher than those of clopidogrel 75 mg/day. CYP2C19 polymorphisms may have reduced clopidogrel-induced IPA.


Assuntos
Citocromo P-450 CYP2C19/genética , Inibidores da Agregação Plaquetária , Agregação Plaquetária/efeitos dos fármacos , Polimorfismo Genético , Cloridrato de Prasugrel , Acidente Vascular Cerebral , Adulto , Idoso , Clopidogrel/administração & dosagem , Clopidogrel/farmacocinética , Citocromo P-450 CYP2C19/administração & dosagem , Citocromo P-450 CYP2C19/farmacocinética , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Cloridrato de Prasugrel/administração & dosagem , Cloridrato de Prasugrel/farmacocinética , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/genética
16.
BMC Neurol ; 19(1): 291, 2019 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-31735164

RESUMO

BACKGROUND: Mutations of cyclooxygenase gene (COX gene) may increase the susceptibility of ischemic stroke. We investigated five variants (rs5788, rs1330344, rs3842788, rs20417, and rs689466) of two COX genes in order to explaining the association between these polymorphisms and we also investigated the association between these variants and ischemic stroke risk to determine whether gene-gene interaction between these genes increases the susceptibility of ischemic stroke or its subtypes. METHODS: A total of 1981 study subjects (1078 cases and 903 control subjects) were recruited. The interaction of multiple factors was investigated using Multifactor Dimensionality Reduction. The additive effect of single nucleotide polymorphisms on ischemic stroke or its subtypes were analyzed by multiple factor logistic regression. RESULTS: At COX-1(rs1330344), AA genotype carriers had a lower susceptibility of ischemic stroke (OR = 0.657, 95%CI = 0.437-0.988, P = 0.044), and A allele carriers had a lower susceptibility of ischemic stroke (OR = 0.812, 95%CI = 0.657-0.978, P = 0.029). At COX-1(rs3842788), AA genotype carriers had a higher susceptibility of ischemic stroke (OR = 5.203, 95% CI = 1.519-5.159, P = 0.016). At COX-2 (rs689466), AA genotype carriers had a higher susceptibility of large-artery atherosclerosis (OR = 1.404, 95% CI = 1.019-1.934, P = 0.038). COX-1(rs1330344, rs3842788) and COX-2 rs689466 interacted in SVO, but had no additive effect with ischemic stroke and other subtypes. CONCLUSIONS: At rs1330344, AA genotype may reduce the susceptibility of ischemic stroke. At rs3842788, AA genotype may increase the susceptibility of ischemic stroke. At rs689466, AA genotype may increase the susceptibility of large-artery atherosclerosis (LAA). COX - 1(rs1330344, rs3842788) and COX-2 rs689466 interacted in small vessel occlusion (SVO), but had no additive effect with ischemic stroke and other subtypes.


Assuntos
Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/genética , Predisposição Genética para Doença/genética , Acidente Vascular Cerebral/genética , Idoso , Povo Asiático/genética , Aterosclerose/enzimologia , Aterosclerose/genética , Isquemia Encefálica/enzimologia , Isquemia Encefálica/genética , Estudos de Casos e Controles , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Acidente Vascular Cerebral/enzimologia
17.
Biosci Rep ; 39(10)2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31652447

RESUMO

BACKGROUND: Birth hypoxia causes neonatal mortality and morbidity. Hypoxia/ischemia can facilitate brain damage, causing various kinds of diseases, such as ischemic stroke. It is necessary to understand the potential underlying mechanisms of ischemic stroke. Previous studies revealed the involvement of thousand and one kinase 1 (TAOK1) in many cellular processes. METHODS: Herein, middle cerebral artery (MCA) occlusion (MCAO) was performed in rats to establish ischemic stroke in the animal model, and cortical neural stem cells from rats were treated with oxygen-glucose deprivation (OGD) to induce ischemic stroke cell model. The animal model of ischemic stroke was validated by Bederson and Zea-Longa neurological deficit scores and rotarod test. TAOK1 expression was examined by quantitative real-time PCR (qRT-PCR), Western blot, and immunofluorescent staining both in vivo and in vitro. RESULT: Compared with sham animals, the MCAO rats showed a significant increase in the neurological scores, and obvious motor behavioral deficits. Meanwhile, there was increased apoptosis and inflammatory response in the model group. TAOK1 overexpression reversed the OGD-induced cell injury, while TAOK1 knockdown exhibited the opposing effects. On the mechanism, the OGD-induced suppression of PI3K/AKT, and activation of mitogen-activated protein kinase (MAPK) signaling pathways were abolished by TAOK1 overexpression, and aggravated by TAOK1 knockdown in vitro. Moreover, we proved that the inhibitory effect of TAOK1 on OGD-induced apoptosis was dependent on the intracellular kinase activity. CONCLUSION: TAOK1 protected MCAO-induced cerebral ischemic stroke by decreasing the pro-inflammatory factors and apoptosis via PI3K/AKT and MAPK signaling pathways.


Assuntos
Apoptose , Isquemia Encefálica/enzimologia , Regulação Enzimológica da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Proteínas Serina-Treonina Quinases/biossíntese , Acidente Vascular Cerebral/enzimologia , Animais , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Inflamação/metabolismo , Inflamação/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/patologia
18.
Eur J Pharmacol ; 861: 172617, 2019 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-31430457

RESUMO

Mitochondrial dysfunctions contribute to brain injury in ischemic stroke while disturbance of mitochondrial dynamics results in mitochondrial dysfunction. Mitochondrial E3 ubiquitin ligase 1 (Mul1) involves in regulation of mitochondrial fission and fusion. This study aims to explore whether Mul1 contributes to brain injury in ischemic stroke and the underlying mechanisms. First, a rat ischemic stroke model was established by middle cerebral artery occlusion (MCAO), which showed ischemic injuries (increase in neurological deficit score and infarct volume) and upregulation of Mul1 in brain tissues. Next, Mul1 siRNAs were injected intracerebroventricularly to knockdown Mul1 expression, which evidently attenuated brain injuries (decrease in neurological deficit score, infarct volume and caspase-3 activity), restored mitochondrial dynamics and functions (decreases in mitochondrial fission and cytochrome c release while increase in ATP production), and restored protein levels of dynamin-related protein 1 (Drp1, a mitochondrial fission protein) and mitofusin2 (Mfn2, a mitochondrial fusion protein) through suppressing their sumoylation and ubiquitination, respectively. Finally, PC12 cells were cultured under hypoxic condition to mimic the ischemic stroke. Consistently, knockdown of Mul1 significantly reduced hypoxic injuries (decrease in apoptosis and LDH release), restored protein levels of Drp1 and Mfn2, recovered mitochondrial dynamics and functions (decreases in mitochondrial fission, mitochondrial membrane potential, reactive oxygen species production and cytochrome c release while increase in ATP production). Based on these observations, we conclude that upregulation of Mul1 contributes to brain injury in ischemic stroke rats and disturbs mitochondrial dynamics through sumoylation of Drp1 and ubiquitination of Mfn2.


Assuntos
Isquemia Encefálica/complicações , Encéfalo/patologia , Dinâmica Mitocondrial , Proteínas Mitocondriais/metabolismo , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Apoptose , Hipóxia Celular , Modelos Animais de Doenças , Dinaminas/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Técnicas de Silenciamento de Genes , Masculino , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Células PC12 , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/enzimologia , Sumoilação , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética , Regulação para Cima
19.
J Cereb Blood Flow Metab ; 39(12): 2379-2391, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31423931

RESUMO

Recent works highlight the therapeutic potential of targeting cyclic guanosine monophosphate (cGMP)-dependent pathways in the context of brain ischemia/reperfusion injury (IRI). Although cGMP-dependent protein kinase I (cGKI) has emerged as a key mediator of the protective effects of nitric oxide (NO) and cGMP, the mechanisms by which cGKI attenuates IRI remain poorly understood. We used a novel, conditional cGKI knockout mouse model to study its role in cerebral IRI. We assessed neurological deficit, infarct volume, and cerebral perfusion in tamoxifen-inducible vascular smooth muscle cell-specific cGKI knockout mice and control animals. Stroke experiments revealed greater cerebral infarct volume in smooth muscle cell specific cGKI knockout mice (males: 96 ± 16 mm3; females: 93 ± 12 mm3, mean±SD) than in all control groups: wild type (males: 66 ± 19; females: 64 ± 14), cGKI control (males: 65 ± 18; females: 62 ± 14), cGKI control with tamoxifen (males: 70 ± 8; females: 68 ± 10). Our results identify, for the first time, a protective role of cGKI in vascular smooth muscle cells during ischemic stroke injury. Moreover, this protective effect of cGKI was found to be independent of gender and was mediated via improved reperfusion. These results suggest that cGKI in vascular smooth muscle cells should be targeted by therapies designed to protect brain tissue against ischemic stroke.


Assuntos
Infarto Cerebral/enzimologia , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Traumatismo por Reperfusão/enzimologia , Acidente Vascular Cerebral/enzimologia , Animais , Infarto Cerebral/genética , Infarto Cerebral/patologia , Proteína Quinase Dependente de GMP Cíclico Tipo I/genética , Feminino , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia
20.
Pathol Res Pract ; 215(9): 152519, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31272760

RESUMO

Ischemic stroke is the main cause of disability and mortality worldwide. Apoptosis and inflammation have an important role in ischemic brain injury. Mesenchymal stem cells (MSCs) have protective effects on stroke treatment due to anti-inflammatory properties. The inhibition of the C-Jun N-terminal kinase (JNK) pathway may be one of the molecular mechanisms of the neuroprotective effect of MSCs in ischemic brain injury. Twenty-eight male Wistar rats were divided randomly into 3 groups. Except the sham group, others subjected to transient middle cerebral artery occlusion (tMCAO). Bone marrow MSCs or saline were injected 3 h after tMCAO. Sensorimotor behavioral tests were performed 24 and 72 h after ischemia and reperfusion (I/R). The rats were sacrificed 72 h after I/R and infarct volume was measured by TTC staining. The number of apoptotic neurons and astrocytes in the peri-infarct area was assessed by TUNEL assay. The morphology of cells was checked by Nissl staining, and the expression of p-JNK was detected by immunohistochemistry and Western blot. Behavioral scores were improved and infarct volume was reduced by MSCs 24 h and 72 h after tMCAO. TUNEL assay showed that neuronal apoptosis and astroglial activity in the penumbra region were reduced by MSCs. Also, Nissl staining showed lower neuronal apoptosis in BMSCs-treated rats compared to controls. JNK phosphorylation which was profoundly induced by ischemia was significantly decreased after MSCs treatment. We concluded that anti-apoptotic and anti-inflammatory effects of MSCs therapy after brain ischemia may be associated with the down-regulation of p-JNK.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Transplante de Células-Tronco Mesenquimais/métodos , Acidente Vascular Cerebral/patologia , Animais , Modelos Animais de Doenças , Masculino , Ratos , Ratos Wistar , Acidente Vascular Cerebral/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...