Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
FASEB J ; 35(11): e21942, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34670018

RESUMEN

Atherosclerosis is a chronic inflammatory disease. Pathophysiological similarities between chronic infections and atherosclerosis triggered interests between these conditions. The seroepidemiological study showed that Helicobacter pylori strains that express cytotoxin-associated gene A (CagA), an oncoprotein and a major virulence factor, was positively correlated with atherosclerosis and related clinical events. Nevertheless, the underlying mechanism is poorly understood. In this study, the seroprevalence of infection by H. pylori and by strains express CagA assessed by enzyme-linked immunosorbent assay (ELISA) showed that the prevalence of CagA strains rather than H. pylori in patients was positively correlated with atherogenesis. Correspondingly, we found that CagA augmented the growth of plaque of ApoE-/- mice in the early stage of atherosclerosis and promoted the expression of adhesion molecules and inflammatory cytokines in mouse aortic endothelial cells (MAECs). Mechanistically, both si-NLRP3 and si-IL-1ß mitigated the promoting effect of CagA on the inflammatory activation of HAECs. In vivo, the inhibition of NLRP3 by MCC950 significantly attenuated the promoting effect of CagA on plaque growth of ApoE-/- mice. We also propose NLRP3 as a potential therapeutic target for CagA-positive H. pylori infection-related atherosclerosis and emphasize the importance of inflammation in atherosclerosis pathology.


Asunto(s)
Antígenos Bacterianos/metabolismo , Aorta/patología , Aterosclerosis/sangre , Proteínas Bacterianas/metabolismo , Caspasa 1/metabolismo , Células Endoteliales/metabolismo , Infecciones por Helicobacter/sangre , Helicobacter pylori/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Placa Aterosclerótica/sangre , Anciano , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Aorta/metabolismo , Aterosclerosis/microbiología , Proteínas Bacterianas/inmunología , Modelos Animales de Enfermedad , Femenino , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/inmunología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Persona de Mediana Edad , Placa Aterosclerótica/microbiología , Estudios Seroepidemiológicos , Células THP-1
2.
Sci Rep ; 11(1): 6177, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33731740

RESUMEN

The purpose of this study was to investigate whether baseline neutrophil to lymphocyte ratio (NLR) was an independent predictor for early symptomatic intracranial hemorrhage (sICH), poor functional outcome and mortality at 3 months after reperfusion therapy in acute ischemic stroke (AIS) patients. Using PubMed and EMBASE, we searched for literature published before January 19th, 2019. Two reviewers independently confirmed each study's eligibility, assessed risk of bias, and extracted data. One reviewer combined studies using random effects meta-analysis. 9 studies with 3651 patients were pooled in the meta-analysis. Overall, baseline NLR levels were greater in patients with poor outcome. The standardized mean difference (SMD) in the NLR levels between patients with poor functional outcome (mRS > 2) and good functional outcome (mRS ≤ 2) was 0.54 units (95% credible interval [CI] [0.38, 0.70]). Heterogeneity test showed that there were significant differences between individual studies (p = 0.02; I2 = 72.8%). The NLR levels were associated with sICH in four included studies (n = 2003, SMD = 0.78, 95% [CI] [0.18, 1.38], I2 = 73.9%). Higher NLR levels were positively correlated with 3-month mortality (n = 1389, ES = 1.71, 95% CI [1.01,2.42], p < 0.01, I2 = 0%) when data were used as categorical variables. Our meta-analysis suggests that increased NLR levels are positively associated with greater risk of sICH, 3-month poor functional outcome and 3-month mortality in AIS patients undergoing reperfusion treatments. Although there are some deficits in this study, it may be feasible to predict the prognosis of reperfusion therapy in AIS patients with NLR levels.


Asunto(s)
Isquemia Encefálica/terapia , Accidente Cerebrovascular Isquémico/epidemiología , Daño por Reperfusión/epidemiología , Reperfusión/efectos adversos , Recuento de Células , Estudios de Cohortes , Humanos , Linfocitos/citología , Neutrófilos/citología , Pronóstico , Factores de Riesgo
3.
Aging (Albany NY) ; 13(4): 5650-5673, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33589571

RESUMEN

Inappropriate activation or overactivation of cyclic GMP-AMP synthase (cGAS) by double-stranded deoxyribonucleic acid (dsDNA) initiates a regulatory signaling cascade triggering a variety of inflammatory responses, which are a great threat to human health. This study focused on identifying the role of cGAS in atherosclerosis and its potential mechanisms. The relationship between cGAS and atherosclerosis was identified in an ApoE -/- mouse model. Meanwhile, RNA sequencing (RNA-seq) analysis of the underlying mechanisms of atherosclerosis in RAW264.7 macrophages treated with cGAS inhibition was conducted. Results showed that cGAS was positively correlated with atherosclerotic plaque area, and was mainly distributed in macrophages. RNA-seq analysis revealed that inflammatory response, immune response and cytokine-cytokine receptor interaction may play important roles in the development of atherosclerosis. Real-time quantitative polymerase chain reaction (RT-qPCR) results showed that the expression of the pro-inflammatory factors, signal transducer and activator of transcription (Stat), interferon regulatory factor (Irf), toll-like receptors (Tlrs), and type I interferons (Ifns) were synergistically reduced when cGAS was inhibited. Furthermore, cGAS inhibition significantly inhibited RAW264.7 macrophage M1 polarization. These results demonstrate that cGAS may contribute to the development of atherosclerosis through synergistic inflammatory signaling of TLRs, STAT/IRF as well as IFNs, leading to macrophage M1 polarization.


Asunto(s)
Aterosclerosis/etiología , Nucleotidiltransferasas/metabolismo , Animales , Aterosclerosis/metabolismo , ADN Mitocondrial/metabolismo , Masculino , Ratones , Ratones Noqueados para ApoE , Mapas de Interacción de Proteínas , Células RAW 264.7 , Transcriptoma
4.
EMBO Mol Med ; 12(2): e10154, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31943789

RESUMEN

Diabetic retinopathy (DR) is a common complication of diabetes and leads to blindness. Anti-VEGF is a primary treatment for DR. Its therapeutic effect is limited in non- or poor responders despite frequent injections. By performing a comprehensive analysis of the semaphorins family, we identified the increased expression of Sema4D during oxygen-induced retinopathy (OIR) and streptozotocin (STZ)-induced retinopathy. The levels of soluble Sema4D (sSema4D) were significantly increased in the aqueous fluid of DR patients and correlated negatively with the success of anti-VEGF therapy during clinical follow-up. We found that Sema4D/PlexinB1 induced endothelial cell dysfunction via mDIA1, which was mediated through Src-dependent VE-cadherin dysfunction. Furthermore, genetic disruption of Sema4D/PlexinB1 or intravitreal injection of anti-Sema4D antibody reduced pericyte loss and vascular leakage in STZ model as well as alleviated neovascularization in OIR model. Moreover, anti-Sema4D had a therapeutic advantage over anti-VEGF on pericyte dysfunction. Anti-Sema4D and anti-VEGF also conferred a synergistic therapeutic effect in two DR models. Thus, this study indicates an alternative therapeutic strategy with anti-Sema4D to complement or improve the current treatment of DR.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforinas/metabolismo , Transducción de Señal , Animales , Antígenos CD , Diabetes Mellitus , Retinopatía Diabética/inducido químicamente , Humanos , Ratones , Neovascularización Patológica
5.
J Mol Cell Cardiol ; 135: 40-51, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31352044

RESUMEN

BACKGROUND: Seroepidemiological studies have highlighted a positive relation between CagA-positive Helicobacter pylori (H. pylori), atherosclerosis and related clinic events. However, this link has not been well validated. The present study was designed to explore the role of H. pylori PMSS1 (a CagA-positive strain that can translocate CagA into host cells) and exosomal CagA in the progression of atherosclerosis. METHODS: To evaluate whether H. pylori accelerates or even induces atherosclerosis, H. pylori-infected C57/BL6 mice and ApoE-/- mice were maintained under different dietary conditions. To identify the role of H. pylori-infected gastric epithelial cells-derived exosomes (Hp-GES-EVs) and exosomal CagA in atherosclerosis, ApoE-/- mice were given intravenous or intraperitoneal injections of saline, GES-EVs, Hp-GES-EVs, and recombinant CagA protein (rCagA). FINDINGS: CagA-positive H. pylori PMSS1 infection does not induce but promotes macrophage-derived foam cell formation and augments atherosclerotic plaque growth and instability in two animal models. Meanwhile, circulating Hp-GES-EVs are taken up in aortic plaque, and CagA is secreted in Hp-GES-EVs. Furthermore, the CagA-containing EVs and rCagA exacerbates macrophage-derived foam cell formation and lesion development in vitro and in vivo, recapitulating the pro-atherogenic effects of CagA-positive H. pylori. Mechanistically, CagA suppresses the transcription of cholesterol efflux transporters by downregulating the expression of transcriptional factors PPARγ and LXRα and thus enhances foam cell formation. INTERPRETATION: These results may provide new insights into the role of exosomal CagA in the pathogenesis of CagA-positive H. pylori infection-related atherosclerosis. It is suggested that preventing and eradicating CagA-positive H. pylori infection could reduce the incidence of atherosclerosis and related events.


Asunto(s)
Antígenos Bacterianos/metabolismo , Aterosclerosis/metabolismo , Proteínas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Células Espumosas/metabolismo , Mucosa Gástrica/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Animales , Aterosclerosis/microbiología , Aterosclerosis/patología , Células Epiteliales/microbiología , Células Epiteliales/patología , Células Espumosas/microbiología , Células Espumosas/patología , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Ratones
6.
Cell Death Dis ; 10(7): 487, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31221990

RESUMEN

Endothelium (EC) is a key component of blood-brain barrier (BBB), and has an important position in the neurovascular unit. Its dysfunction and death after cerebral ischemic/reperfusion (I/R) injury not only promote evolution of neuroinflammation and brain edema, but also increase the risk of intracerebral hemorrhage of thrombolytic therapies. However, the mechanism and specific interventions of EC death after I/R injury are poorly understood. Here we showed that necroptosis was a mechanism underlying EC death, which promoted BBB breakdown after I/R injury. Treatment of rats with receptor interacting protein kinase 1 (RIPK1)-inhibitor, necrostatin-1 reduced endothelial necroptosis and BBB leakage. We furthermore showed that perivascular M1-like microglia-induced endothelial necroptosis leading to BBB disruption requires tumor necrosis factor-α (TNF-α) secreted by M1 type microglia and its receptor, TNF receptor 1 (TNFR1), on endothelium as the primary mediators of these effects. More importantly, anti-TNFα (infliximab, a potent clinically used drug) treatment significantly ameliorate endothelial necroptosis, BBB destruction and improve stroke outcomes. Our data identify a previously unexplored role for endothelial necroptosis in BBB disruption and suggest infliximab might serve as a potential drug for stroke therapy.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Necroptosis/fisiología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Western Blotting , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Etiquetado Corte-Fin in Situ , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Microscopía Electrónica de Transmisión , Necroptosis/genética , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/genética
7.
Cell Death Dis ; 10(2): 138, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755588

RESUMEN

Atherosclerotic cardio-cerebrovascular disease and death remain the leading cause of morbidity and mortality worldwide. Defective efferocytosis, the clearance of apoptotic cells by macrophages, is thought to lead to increased inflammation and necrotic core formation in atherosclerotic lesions. However, very little is known about the role of long noncoding RNA (lncRNA) during this process. Here we show that lncRNA myocardial infarction associated transcript (MIAT) was markedly elevated in the serum of patients with symptoms of vulnerable atherosclerotic plaque and the macrophages of necrotic cores in an advanced atherosclerosis mouse model. MIAT knockdown attenuated atherosclerosis progression, reduced necrotic core size, and increased plaque stability in vivo. Furthermore, MIAT knockdown promoted clearance of apoptotic cells by macrophages in vivo and in vitro. Mechanistic studies revealed that MIAT acted as a micro RNA (miRNA) sponge to positively modulate the expression of anti-phagocytic molecule CD47 through sponging miR-149-5p. Together, these findings identified a macrophage MIAT/miR-149-5p /CD47 pathway as a key factor in the development of necrotic atherosclerotic plaques.


Asunto(s)
Antígeno CD47/metabolismo , Enfermedades de las Arterias Carótidas/sangre , MicroARNs/metabolismo , Fagocitosis/genética , ARN Largo no Codificante/sangre , Regulación hacia Arriba , Anciano , Animales , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados para ApoE , MicroARNs/genética , Persona de Mediana Edad , Células RAW 264.7 , ARN Largo no Codificante/genética , Transfección
8.
FASEB J ; 33(4): 4947-4961, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30653356

RESUMEN

Angiogenesis is a crucial defense response to hypoxia that regulates the process of raising the promise of long-term neurologic recovery during the management of stroke. A high expression of antiangiogenic factors leads to the loss of neovascularization capacity in pathologic conditions. We have previously documented an impairment of the cerebral vessel perfusion and neovascularization in the cortex neighboring the stroke-induced lesion, which was accompanied by an activation of semaphorin 3E (Sema3E)/PlexinD1 after ischemic stroke. In this study, we employed micro-optical sectioning tomography to fully investigate the details of the vascular pattern, including the capillaries. We found that after transient middle cerebral artery occlusion, inhibiting PlexinD1 signaling led to an organized recovery of the vascular network in the ischemic area. We then further explored the possible mechanisms. In vivo, Sema3E substantially decreased dynamic delta-like 4 (DLL4) expression. In cultured brain microvascular endothelial cells, Sema3E down-regulated DLL4 expression via inhibiting Ras-related C3 botulinum toxin substrate 1-induced JNK phosphorylation. At the microcosmic level, Sema3E/PlexinD1 signaling promoted F-actin disassembly and focal adhesion reduction by activating the small guanosine triphosphatase Ras homolog family member J by releasing RhoGEF Tuba from direct binding to PlexinD1, thus mediating endothelial cell motility and filopodia retraction. Our study reveals that Sema3E/PlexinD1 signaling, which suppressed endothelial DLL4 expression, cell motility, and filopodia formation, is expected to be a novel druggable target for angiogenesis during poststroke progression.-Zhou, Y.-F., Chen, A.-Q., Wu, J.-H., Mao, L., Xia, Y.-P., Jin, H.-J., He, Q.-W., Miao, Q. R., Yue, Z.-Y., Liu, X.-L., Huang, M., Li, Y.-N., Hu, B. Sema3E/PlexinD1 signaling inhibits postischemic angiogenesis by regulating endothelial DLL4 and filopodia formation in a rat model of ischemic stroke.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Seudópodos/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforinas/metabolismo , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Células Cultivadas , Células Endoteliales/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Feocromocitoma/metabolismo , Feocromocitoma/patología , Seudópodos/genética , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/genética , Semaforinas/genética , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
9.
EBioMedicine ; 39: 95-108, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30579864

RESUMEN

BACKGROUND: Neointimal hyperplasia is a prominent pathological event during in-stent restenosis. Phenotype switching of vascular smooth muscle cells (VSMCs) from a differentiated/contractile to a dedifferentiated/synthetic phenotype, accompanied by migration and proliferation of VSMCs play an important role in neointimal hyperplasia. However, the molecular mechanisms underlying phenotype switching of VSMCs have yet to be fully understood. METHODS: The mouse carotid artery ligation model was established to evaluate Sema3A expression and its role during neointimal hyperplasia in vivo. Bioinformatics analysis, chromatin immunoprecipitation (ChIP) assays and promoter-luciferase reporter assays were used to examine regulatory mechanism of Sema3A expression. SiRNA transfection and lentivirus infection were performed to regulate Sema3A expression. EdU assays, Wound-healing scratch experiments and Transwell migration assays were used to assess VSMC proliferation and migration. FINDINGS: In this study, we found that semaphorin-3A (Sema3A) was significantly downregulated in VSMCs during neointimal hyperplasia after vascular injury in mice and in human atherosclerotic plaques. Meanwhile, Sema3A was transcriptionally downregulated by PDGF-BB via p53 in VSMCs. Furthermore, we found that overexpression of Sema3A inhibited VSMC proliferation and migration, as well as increasing differentiated gene expression. Mechanistically, Sema3A increased the NRP1-plexin-A1 complex and decreased the NRP1-PDGFRß complex, thus inhibiting phosphorylation of PDGFRß. Moreover, we found that overexpression of Sema3A suppressed neointimal hyperplasia after vascular injury in vivo. INTERPRETATION: These results suggest that local delivery of Sema3A may act as a novel therapeutic option to prevent in-stent restenosis.


Asunto(s)
Aterosclerosis/genética , Neointima/prevención & control , Semaforina-3A/genética , Lesiones del Sistema Vascular/genética , Animales , Aterosclerosis/metabolismo , Becaplermina/metabolismo , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Humanos , Ratones , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Neointima/genética , Neointima/metabolismo , Semaforina-3A/metabolismo , Transducción de Señal , Transcripción Genética , Lesiones del Sistema Vascular/metabolismo
10.
J Mol Cell Cardiol ; 123: 139-149, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30227118

RESUMEN

Disruption of carotid vulnerable atherosclerotic plaque is responsible for acute ischemic stroke (AIS) and the early detection and intervention approach are greatly limited. Undertaking a microarray of microRNAs (miRNAs) in the plasma of AIS patients with carotid vulnerable plaques, miR-23a-5p was markedly elevated and was positively correlated with the plaque progression and vulnerability. Correspondingly, we found that miR-23a-5p expression was significantly increased in both plasma and macrophages from atherosclerosis mice. Bioinformatics analysis and in vitro knockdown experiments identified that ATP-binding cassette transporter A1/G1 as a novel target of miR-23a-5p. Luciferase reporter assays showed that miR-23a-5p repressed the 3' untranslated regions (UTR) activity of ABCA1/G1. Moreover, functional analyses demonstrated that transfection of miR-23a-5p inhibitor enhanced cholesterol efflux and decreased foam cell formation through upregulating ABCA1/G1 expression levels. Furthermore, long term in vivo systemically delivered miR-23a-5p antagomir significantly increased ABCA1/G1 expression in the aorta of ApoE-/- mice. Importantly, the miR-23a-5p antagomir therapy significantly reduced atherosclerosis progression and promoted plaque stability. Our observations indicate that miR-23a-5p promotes macrophage-derived foam cell formation and might be a key regulator contributing to atherosclerotic plaque progression and vulnerability.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Macrófagos/metabolismo , MicroARNs/genética , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Regiones no Traducidas 3' , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Placa Aterosclerótica/patología , Células RAW 264.7 , Interferencia de ARN
11.
Trop Med Int Health ; 23(11): 1200-1206, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30178470

RESUMEN

OBJECTIVES: A good mastery of stroke-related knowledge can be of great benefit in developing healthy behaviours. This study surveyed the knowledge about stroke and influencing factors among patients with acute ischaemic stroke (AIS) at discharge in a Chinese province. METHODS: A cross-section study was conducted from November 1, 2014 to January 31, 2015. A total of 1531 AIS patients in Hubei Province completed a questionnaire at discharge. Multivariate linear regression was used to identify the influencing factors of their knowledge of stroke. RESULTS: About 31.2% of the respondents did not know that stroke is caused by blockage or rupture of cerebral blood vessels and 20.3% did not realise they need immediate medical attention after onset. Approximately 50% did not know that sudden blurred vision, dizziness, headache and unconsciousness are the warning signs of stroke. Over 40% were not aware of the risk factors of the condition, such as hypertension, hyperlipidaemia, diabetes mellitus, smoking and obesity. Over 20% had no idea that they need long-term medication and strict control of blood pressure, blood lipids and blood sugar. Their knowledge levels were correlated with regions of residence (P < 0.0001), socioeconomic status (P < 0.05), physical condition (P < 0.01), previous stroke (P < 0.0001) and family members and friends having had a stroke (P < 0.01). CONCLUSIONS: Most AIS patients in Hubei Province, China, had little knowledge of stroke at discharge. Further efforts should be devoted to strengthening the in-hospital education of stroke patients, especially those with a low income and those from rural areas.


Asunto(s)
Conocimientos, Actitudes y Práctica en Salud , Alfabetización en Salud/estadística & datos numéricos , Alta del Paciente , Accidente Cerebrovascular/fisiopatología , Adulto , Anciano , Anciano de 80 o más Años , China , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo , Encuestas y Cuestionarios
12.
Neurobiol Aging ; 70: 102-116, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30007159

RESUMEN

Brain tissue survival and functional recovery after ischemic stroke greatly depend on cerebral vessel perfusion and functional collateral circulation in the ischemic area. Semaphorin 3E (Sema3E), one of the class 3 secreted semaphorins, has been demonstrated to be a critical regulator in embryonic and postnatal vascular formation via binding to its receptor PlexinD1. However, whether Sema3E/PlexinD1 signaling is involved in poststroke neovascularization remains unknown. To determine the contribution of Sema3E/PlexinD1 signaling to poststroke recovery, aged rats (18 months) were subjected to a transient middle cerebral artery occlusion. We found that depletion of Sema3E/PlexinD1 signaling with lentivirus-mediated PlexinD1-specific-shRNA improves tissue survival and functional outcome. Sema3E/PlexinD1 inhibition not only increases cortical perfusion but also ameliorates blood-brain barrier damage, as determined by positron emission tomography and magnetic resonance imaging. Mechanistically, we demonstrated that Sema3E suppresses endothelial cell proliferation and angiogenic capacity. More importantly, Sema3E/PlexinD1 signaling inhibits recruitment of pericytes by decreasing production of platelet derived growth factor-BB in endothelial cells. Overall, our study revealed that inhibition of Sema3E/PlexinD1 signaling in the ischemic penumbra, which increases both endothelial angiogenic capacity and recruitment of pericytes, contributed to functional neovascularization and blood-brain barrier integrity in the aged rats. Our findings imply that Sema3E/PlexinD1 signaling is a novel therapeutic target for improving brain tissue survival and functional recovery after ischemic stroke.


Asunto(s)
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforina-3A/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Barrera Hematoencefálica/patología , Encéfalo/patología , Isquemia Encefálica/patología , Masculino , Neovascularización Patológica/fisiopatología , Neuropilina-1/antagonistas & inhibidores , Neuropilina-1/metabolismo , Ratas Sprague-Dawley , Recuperación de la Función , Semaforina-3A/antagonistas & inhibidores , Transducción de Señal , Accidente Cerebrovascular/patología , Regulación hacia Arriba
13.
FASEB J ; 32(6): 3133-3148, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29401609

RESUMEN

Blood-brain barrier (BBB) disruption caused by reperfusion injury after ischemic stroke is an intractable event conducive to further injury. Brain pericytes play a vital role in maintaining BBB integrity by interacting with other components of the BBB. In this study, we found that sphingosine-1-phosphate receptor (S1PR)2 expressed in pericytes was significantly up-regulated after ischemia in vivo and in vitro. By using a S1PR2 antagonist (JTE-013), we showed that S1PR2 plays a critical role in the induction of BBB permeability of transient middle cerebral artery occlusion (tMCAO) rats and the in vitro BBB model. Furthermore, we discovered that S1PR2 may decrease N-cadherin expression and increase pericyte migration via NF-κB p65 signal and found that S1PR2 could be regulated by miR-149-5p negatively, which was decreased in the ischemic boundary zone and cultured pericytes after ischemia. Overexpression of miR-149-5p in cultured pericytes substantially increased N-cadherin expression and decreased pericyte migration, which decreased BBB leakage in the in vitro model. Up-regulating miR-149-5p by intracerebroventricular injection of agomir-149-5p attenuated BBB permeability and improved the outcomes of tMCAO rats significantly. Thus, our data suggest that miR-149-5p may serve as a potential target for treatment of BBB disruption after ischemic stroke.-Wan, Y., Jin, H.-J., Zhu, Y.-Y., Fang, Z., Mao, L., He, Q., Xia, Y.-P., Li, M., Li, Y., Chen, X., Hu, B. MicroRNA-149-5p regulates blood-brain barrier permeability after transient middle cerebral artery occlusion in rats by targeting S1PR2 of pericytes.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Permeabilidad Capilar , MicroARNs/metabolismo , Pericitos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Barrera Hematoencefálica/patología , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Movimiento Celular/genética , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media , Masculino , MicroARNs/genética , Pericitos/patología , Ratas , Ratas Sprague-Dawley , Receptores de Lisoesfingolípidos/genética , Transducción de Señal/genética , Receptores de Esfingosina-1-Fosfato , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
14.
FASEB J ; 32(2): 935-944, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29070584

RESUMEN

Blood-brain barrier (BBB) disruption plays a critical role in brain injury induced by cerebral ischemia, and preserving BBB integrity during ischemia could alleviate cerebral injury. We examined the role of miR-130a in ischemic BBB disruption by using models of rat middle cerebral artery occlusion and cell oxygen-glucose deprivation. We found that ischemia significantly increased microRNA-130a (miR-130a) level and that miR-130a was predominantly from brain microvascular endothelial cells. Antagomir-130a, an antagonist of miR-130a, could attenuate brain edema, lower BBB permeability, reduce infarct volume, and improve neurologic function. MiR-130a overexpression induced by miR-130a mimic increased monolayer permeability, and intercellular inhibition of miR-130a by a miR-130a inhibitor suppressed oxygen-glucose deprivation-induced increase in monolayer permeability. Moreover, dual luciferase reporter system showed that Homeobox A5 was the direct target of miR-130a. MiR-130a, by inhibiting Homeobox A5 expression, could down-regulate occludin, thereby increasing BBB permeability. Our results suggested that miR-130a might be implicated in ischemia-induced BBB dysfunction and serve as a target for the treatment of ischemic stroke.-Wang, Y., Wang, M.-D., Xia, Y.-P., Gao, Y., Zhu, Y.-Y., Chen, S.-C., Mao, L., He, Q.-W., Yue, Z.-Y., Hu, B. MicroRNA-130a regulates cerebral ischemia-induced blood-brain barrier permeability by targeting Homeobox A5.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Proteínas de Homeodominio/metabolismo , MicroARNs/metabolismo , Ocludina/biosíntesis , Animales , Barrera Hematoencefálica/patología , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Proteínas de Homeodominio/genética , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Ocludina/genética , Oligonucleótidos/farmacología , Permeabilidad , Ratas , Ratas Sprague-Dawley
15.
Sci Rep ; 7(1): 14926, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29097744

RESUMEN

Fluoxetine, one of the selective serotonin reuptake inhibitor (SSRI) antidepressants, has been thought to be effective for treating post-stroke depression (PSD). Recent work has shown that fluoxetine may exert an antidepressive effect through increasing the level of brain-derived neurotrophic factor (BDNF), but the underlying mechanism still remains unclear. In the present study, we successfully established the PSD model using male C57BL/6 J mice by photothrombosis of the left anterior cortex combined with isolatied-housing conditions. In the process, we confirmed that fluoxetine could improve the depression-like behaviors of PSD mice and upregulate the expression of BDNF in the hippocampus. However, depletion of BDNF by transfecting lentivirus-derived shBDNF in hippocampus suppressed the effect of fluoxetine. Furthermore, we demonstrated the epigenetic mechanisms involved in regulation of BDNF expression induced by fluoxetine. We found a statistically significant increase in DNA methylation at specific CpG sites (loci 2) of Bdnf promoter IV in the hippocampus of PSD mice. We also found that fluoxetine treatment could disassociate the MeCP2-CREB-Bdnf promoter IV complex via phosphorylation of MeCP2 at Ser421 by Protein Kinase A (PKA). Our research highlighted the importance of fluoxetine in regulating BDNF expression which could represent a potential strategy for preventing PSD.


Asunto(s)
Antidepresivos de Segunda Generación/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/genética , Depresión/tratamiento farmacológico , Depresión/etiología , Epigénesis Genética/efectos de los fármacos , Fluoxetina/uso terapéutico , Accidente Cerebrovascular/complicaciones , Animales , Antidepresivos de Segunda Generación/farmacología , Depresión/genética , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/etiología , Trastorno Depresivo/genética , Modelos Animales de Enfermedad , Fluoxetina/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones Endogámicos C57BL , Activación Transcripcional/efectos de los fármacos
16.
Cardiovasc Res ; 113(14): 1763-1775, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29016743

RESUMEN

AIMS: The migration and proliferation of vascular smooth muscle cells (VSMCs) are crucial events in the neointimal formation, a hallmark of atherosclerosis and restenosis. Semaphorin3E (Sema3E) has been found to be a critical regulator of cell migration and proliferation in many scenarios. However, its role on VSMCs migration and proliferation is unclear. This study aimed to investigate the effect of Sema3E on VSMCs migration, proliferation and neointimal formation, and explore possible mechanisms. METHODS AND RESULTS: We found that the expression of Sema3E was progressively decreased during neointimal formation in a carotid ligation model. H&E-staining showed lentivirus-mediated overexpression of Sema3E in carotid ligation area attenuated neointimal formation. Immunofluorescence staining showed that the receptor (PlexinD1) of Sema3E was expressed in vascular walls. In cultured mouse VSMCs, Sema3E inhibited VSMCs migration and proliferation via plexinD1 receptor. The inhibitory effect was mediated, at least in part, by inactivating Rap1-AKT signalling pathways in VSMCs. Moreover, we found that PDGFBB down-regulated the expression of Sema3E in VSMCs and Sema3E notably inhibited the expression of PDGFB in endothelial cells. In addition, the number of Sema3E-positive VSMCs was diminished in plaques of atherosclerotic patients. Results from a public GEO microarray database showed a negative correlation between Sema3E and PDGFB transcriptional levels in the human plaques examined. CONCLUSION: Our study demonstrates that Sema3E/plexinD1 inhibits proliferation and migration of VSMCs via inactivation of Rap1-AKT signalling pathways. The mutual inhibition between PDGF-BB and Sema3E after vascular injury plays a critical role in the process of neointimal formation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Glicoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Neointima/tratamiento farmacológico , Semaforinas/metabolismo , Animales , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Traumatismos de las Arterias Carótidas/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Proteínas del Citoesqueleto , Humanos , Masculino , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Transducción de Señal/efectos de los fármacos
17.
J Huazhong Univ Sci Technolog Med Sci ; 37(3): 433-438, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28585150

RESUMEN

Mild encephalopathy/encephalitis with a reversible splenial (MERS) lesion is a clinic-radiological entity. The clinical features of MERS in neonates are still not systemically reported. This paper presents five cases of MERS, and the up-to-date reviews of previously reported cases were collected and analyzed in the literature. Here we describe five cases clinically diagnosed with MERS. All of them were neonates and the average age was about 4 days. They were admitted for the common neurological symptoms such as hyperspasmia, poor reactivity and delirium. Auxiliary examinations during hospitalization also exhibited features in common. In this report, we reached following conclusions. Firstly, magnetic resonance imaging revealed solitary or comprehensive lesions in the splenium of corpus callosum, some of them extending to almost the whole corpus callosum. The lesions showed low intensity signal on T1-weighted images, homogeneously hyperintense signal on T2-weighted images, fluid-attenuated inversion recovery and diffusion-weighted images, and exhibited an obvious reduced diffusion on apparent diffusion coefficient map. Moreover, the lesions in the magnetic resonance imaging disappeared very quickly even prior to the clinical recovery. Secondly, all the cases depicted here suffered electrolyte disturbances especially hyponatremia which could be easily corrected. Lastly, all of the cases recovered quickly over one week to one month and majority of them exhibited signs of infections and normal electroencephalography.


Asunto(s)
Encefalopatías/diagnóstico por imagen , Cuerpo Calloso/diagnóstico por imagen , Encefalitis/diagnóstico por imagen , Hiponatremia/diagnóstico , Convulsiones/diagnóstico por imagen , Encefalopatías/tratamiento farmacológico , Encefalopatías/fisiopatología , Cefoperazona/uso terapéutico , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/patología , Electroencefalografía , Encefalitis/tratamiento farmacológico , Encefalitis/fisiopatología , Hospitalización , Humanos , Hiponatremia/tratamiento farmacológico , Hiponatremia/fisiopatología , Recién Nacido , Imagen por Resonancia Magnética , Masculino , Fenobarbital/uso terapéutico , Recuperación de la Función/fisiología , Convulsiones/tratamiento farmacológico , Convulsiones/fisiopatología , Cloruro de Sodio/uso terapéutico , Equilibrio Hidroelectrolítico
18.
Neuroscience ; 352: 285-295, 2017 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-28391011

RESUMEN

The Sonic hedgehog (Shh) signaling pathway is recapitulated in response to ischemic injury. Here, we investigated the clinical implications of Shh protein in the ischemic stroke and explored the underlying mechanism. Intracerebroventricular injection of Shh, Cyclopamine, or anti-vascular endothelial growth factor (VEGF) was performed immediately after permanent middle cerebral artery occlusion (pMCAO) surgery and lasted for 7days (d). Phosphate-buffered saline (PBS) was used as control. Neurological deficits and infarct volume were examined 7d after pMCAO. Microvascular density with fluorescein-iso-thiocyanate (FITC) assay and double staining with CD31 and Ki-67 was measured at 7d. To observe in vitro angiogenesis, rat brain microvascular endothelial cells (RBMECs) were incubated under oxygen glucose deprivation (OGD) for 6h (h) and treated with Shh/anti-VEGF. We found that (1) Shh improved neurological scores and reduced infarct volume, which was blocked by Cyclopamine, (2) Shh improved the microvascular density and promoted angiogenesis and neuron survival in the ischemic boundary zone, (3) Shh enhanced VEGF expression and VEGF antibody could reverse angiogenic and protective effect of Shh in vivo and in vitro. These data demonstrate that the administration of Shh protein could protect brain from ischemic injury, in part by promoting angiogenic repair.


Asunto(s)
Inductores de la Angiogénesis/uso terapéutico , Proteínas Hedgehog/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/etiología , Accidente Cerebrovascular/complicaciones , Animales , Encéfalo/anatomía & histología , Infarto Encefálico/etiología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Colecistoquinina/metabolismo , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Dineínas/genética , Dineínas/metabolismo , Células Endoteliales/efectos de los fármacos , Proteínas Hedgehog/metabolismo , Masculino , Receptor Patched-1/genética , Receptor Patched-1/metabolismo , Fragmentos de Péptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Accidente Cerebrovascular/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Biochem Biophys Res Commun ; 485(1): 167-173, 2017 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-28202414

RESUMEN

Emerging studies have illustrated that LncRNAs TUG1 play critical roles in multiple biologic processes. However, the LncRNA TUG1 expression and function in ischemic stroke have not been reported yet. In this study, we found that LncRNA TUG1 expression was significantly up-regulated in brain ischemic penumbra from rat middle carotid artery occlusion (MCAO) model, while similar results were also observed in cultured neurons under oxygen-glucose deprivation (OGD) insult. Knockdown of TUG1 decreased the ratio of apoptotic cells and promoted cells survival in vitro, which may be regulated by the elevated miRNA-9 expression and decreased Bcl2l11 protein. Furthermore, TUG1 could directly interact with miR-9 and down-regulating miR-9 could efficiently reverse the function of TUG1 on the Bcl2l11 expression. In summary, our result sheds light on the role of LncRNA TUG1 as a miRNA sponge for ischemic stroke, possibly providing a new therapeutic target in stroke.


Asunto(s)
Apoptosis , Proteína 11 Similar a Bcl2/genética , Isquemia Encefálica/genética , Encéfalo/patología , Regulación de la Expresión Génica , MicroARNs/genética , ARN Largo no Codificante/genética , Animales , Encéfalo/metabolismo , Isquemia Encefálica/patología , Células Cultivadas , Técnicas de Silenciamiento del Gen , Masculino , Neuronas/metabolismo , Neuronas/patología , Ratas Sprague-Dawley , Regulación hacia Arriba
20.
Arterioscler Thromb Vasc Biol ; 37(3): 515-524, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28062501

RESUMEN

OBJECTIVE: P2Y12 is a well-recognized receptor expressed on platelets and the target of thienopyridine-type antiplatelet drugs. However, recent evidence suggests that P2Y12 expressed in vessel wall plays a role in atherogenesis, but the mechanisms remain elusive. In this study, we examined the molecular mechanisms of how vessel wall P2Y12 mediates vascular smooth muscle cells (VSMCs) migration and promotes the progression of atherosclerosis. APPROACH AND RESULTS: Using a high-fat diet-fed apolipoprotein E-deficient mice model, we found that the expression of P2Y12 in VSMCs increased in a time-dependent manner and had a linear relationship with the plaque area. Moreover, administration of P2Y12 receptor antagonist for 12 weeks caused significant reduction in atheroma and decreased the abundance of VSMCs in plaque. In cultured VSMCs, we found that activation of P2Y12 receptor inhibited cAMP/protein kinase A signaling pathway, which induced cofilin dephosphorylation and filamentous actin disassembly, thereby enhancing VSMCs motility and migration. In addition, the number of P2Y12-positive VSMCs was decreased in the carotid artery plaque from patients receiving clopidogrel. CONCLUSIONS: Vessel wall P2Y12 receptor, which promotes VSMCs migration through cofilin dephosphorylation, plays a critical role in the development of atherosclerotic lesion and may be used as a therapeutic target for atherosclerosis.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Movimiento Celular , Cofilina 2/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Clopidogrel , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Fenotipo , Fosforilación , Placa Aterosclerótica , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico , Interferencia de ARN , Receptores Purinérgicos P2Y12/efectos de los fármacos , Receptores Purinérgicos P2Y12/genética , Transducción de Señal , Ticlopidina/análogos & derivados , Ticlopidina/uso terapéutico , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...