Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 41(9): 2399-2416, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34289702

RESUMO

Objective: Vascular smooth muscle cell (VSMC) plasticity plays a critical role in the development of atherosclerosis. Long noncoding RNAs (lncRNAs) are emerging as important regulators in the vessel wall and impact cellular function through diverse interactors. However, the role of lncRNAs in regulating VSMCs plasticity and atherosclerosis remains unclear. Approach and Results: We identified a VSMC-enriched lncRNA cardiac mesoderm enhancer-associated noncoding RNA (CARMN) that is dynamically regulated with progression of atherosclerosis. In both mouse and human atherosclerotic plaques, CARMN colocalized with VSMCs and was expressed in the nucleus. Knockdown of CARMN using antisense oligonucleotides in Ldlr−/− mice significantly reduced atherosclerotic lesion formation by 38% and suppressed VSMCs proliferation by 45% without affecting apoptosis. In vitro CARMN gain- and loss-of-function studies verified effects on VSMC proliferation, migration, and differentiation. TGF-ß1 (transforming growth factor-beta) induced CARMN expression in a Smad2/3-dependent manner. CARMN regulated VSMC plasticity independent of the miR143/145 cluster, which is located in close proximity to the CARMN locus. Mechanistically, lncRNA pulldown in combination with mass spectrometry analysis showed that the nuclear-localized CARMN interacted with SRF (serum response factor) through a specific 600­1197 nucleotide domain. CARMN enhanced SRF occupancy on the promoter regions of its downstream VSMC targets. Finally, knockdown of SRF abolished the regulatory role of CARMN in VSMC plasticity. Conclusions: The lncRNA CARMN is a critical regulator of VSMC plasticity and atherosclerosis. These findings highlight the role of a lncRNA in SRF-dependent signaling and provide implications for a range of chronic vascular occlusive disease states.


Assuntos
Aterosclerose/metabolismo , Plasticidade Celular , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , RNA Longo não Codificante/metabolismo , Fator de Resposta Sérica/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/patologia , Linhagem Celular , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Fenótipo , Placa Aterosclerótica , RNA Longo não Codificante/genética , Receptores de LDL/deficiência , Receptores de LDL/genética , Fator de Resposta Sérica/genética , Transdução de Sinais
2.
Inflammation ; 44(4): 1263-1273, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34076811

RESUMO

MicroRNAs (miRNAs) are short endogenous noncoding RNAs regulating protein translation. However, the specific mechanism by which miR-181b influences sepsis via high-mobility group box-1 protein (HMGB1) still remains unknown. Thus, the aim of this study is to investigate the mechanism of miR-181b in regulating inflammatory response in sepsis-induced myocardial injury through targeting high-mobility group box-1 protein (HMGB1). Through cecal ligation and puncture (CLP), the rat model of sepsis was established. Then, the effect of altered expression of miR-181b and HMGB1 on cardiomyocytes was investigated. The positive expression rate of HMGB1, concentration of inflammatory factors, and serum myocardial enzyme of myocardial tissues were determined. Besides, the binding site between miR-181b and HMGB1 was determined by bioinformatics information and dual-luciferase reporter gene assay. The expression of related genes in cells of each group was determined by RT-qPCR and western blot analysis, and the apoptosis rate of transfected cells in each group was determined by TUNEL assay. HMGB1 expression and inflammatory factors were significantly increased in myocardial tissue of rats with sepsis. Cell morphology and the infiltration of inflammatory cells were significantly improved by overexpression of miR-181b. miR-181b directly targeted HMGB1, and downregulation of HMGB1 reduced inflammatory factors and myocardial injury and inhibited cardiomyocyte apoptosis in sepsis. This present study suggests that miR-181b decreased inflammatory factors and reduced myocardial injury in sepsis through downregulation of HMGB1. Thus, a better understanding of this process may aid in the development of novel therapeutic agents in sepsis.


Assuntos
Regulação para Baixo/fisiologia , Proteína HMGB1/biossíntese , Mediadores da Inflamação/metabolismo , MicroRNAs/biossíntese , Miócitos Cardíacos/metabolismo , Sepse/metabolismo , Animais , Proteína HMGB1/antagonistas & inibidores , Mediadores da Inflamação/antagonistas & inibidores , Masculino , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , Ratos , Ratos Sprague-Dawley , Sepse/patologia
3.
Sci Transl Med ; 12(531)2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-32075942

RESUMO

Long noncoding RNAs (lncRNAs) are emerging regulators of biological processes in the vessel wall; however, their role in atherosclerosis remains poorly defined. We used RNA sequencing to profile lncRNAs derived specifically from the aortic intima of Ldlr -/- mice on a high-cholesterol diet during lesion progression and regression phases. We found that the evolutionarily conserved lncRNA small nucleolar host gene-12 (SNHG12) is highly expressed in the vascular endothelium and decreases during lesion progression. SNHG12 knockdown accelerated atherosclerotic lesion formation by 2.4-fold in Ldlr -/- mice by increased DNA damage and senescence in the vascular endothelium, independent of effects on lipid profile or vessel wall inflammation. Conversely, intravenous delivery of SNHG12 protected the tunica intima from DNA damage and atherosclerosis. LncRNA pulldown in combination with liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis showed that SNHG12 interacted with DNA-dependent protein kinase (DNA-PK), an important regulator of the DNA damage response. The absence of SNHG12 reduced the DNA-PK interaction with its binding partners Ku70 and Ku80, abrogating DNA damage repair. Moreover, the anti-DNA damage agent nicotinamide riboside (NR), a clinical-grade small-molecule activator of NAD+, fully rescued the increases in lesional DNA damage, senescence, and atherosclerosis mediated by SNHG12 knockdown. SNHG12 expression was also reduced in pig and human atherosclerotic specimens and correlated inversely with DNA damage and senescent markers. These findings reveal a role for this lncRNA in regulating DNA damage repair in the vessel wall and may have implications for chronic vascular disease states and aging.


Assuntos
Dano ao DNA , Proteína Quinase Ativada por DNA , Endotélio Vascular/patologia , RNA Longo não Codificante , Animais , Movimento Celular , Proliferação de Células , Cromatografia Líquida , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Proteínas Quinases , RNA Longo não Codificante/genética , Suínos , Espectrometria de Massas em Tandem
4.
Artigo em Inglês | MEDLINE | ID: mdl-29462987

RESUMO

Due to urbanisation, there are large amounts of waste concrete, particularly in rapidly industrialising countries. Currently, demolished concrete is mainly recycled as aggregate for reconstruction. This study has shown that larger sizes (2-5 mm) of recycled concrete aggregate (RCA) removed more than 90% of P from effluent when at pH 5. Analysis of the data, using equilibrium models, indicated a best fit with the Langmuir which predicated an adsorption capacity of 6.88 mg/g. Kinetic analysis indicated the equilibrium adsorption time was 12 h, with pseudo second-order as the best fit. The thermal dynamic tests showed that the adsorption was spontaneous and, together with the evidence from the sequential extraction and desorption experiments, indicated the initial mechanism was physical attraction to the surface followed by chemical reactions which prevented re-release. These results suggested that RCA could be used for both wastewater treatment and P recovery.


Assuntos
Materiais de Construção , Fósforo/química , Reciclagem , Águas Residuárias/química , Poluentes Químicos da Água/química , Purificação da Água/métodos , Adsorção , Cinética
5.
Sci Rep ; 7(1): 12580, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28974760

RESUMO

We developed a virus-like particle (VLP)-based therapeutic vaccine against angiotensin II receptor type 1, ATR-AP205-001, which could significantly reduce the blood pressure and protect target organs of hypertensive animals. In this study, we focused on the immunological effect and safety of the VLP-based vaccine. By comparing to the depolymerized dimeric vaccine ATR-Dimer-001, we found that ATR-AP205-001 reached subcapsular sinus of lymph node shortly after administration, followed by accumulation on follicle dendritic cells via follicle B cell transportation, while ATR-Dimer-001 vaccine showed no association with FDCs. ATR-AP205-001 vaccine strongly activated dendritic cells, which promoted T cells differentiation to follicular helper T cells. ATR-AP205-001 vaccine induced powerful germinal center reaction, which was translated to a boost of specific antibody production and long-lasting B cell memory, far superior to ATR-Dimer-001 vaccine. Moreover, neither cytotoxic T cells, nor Th1/Th17 cell-mediated inflammation was observed in ATR-AP205-001 vaccine, similar to ATR-Dimer-001 vaccine. We concluded that ATR-AP205-001 vaccine quickly induced potent humoral immunity through collaboration of B cells, follicular dendritic cells and follicular helper T cells, providing an effective and safe intervention for hypertension in the future clinical application.


Assuntos
Hipertensão/tratamento farmacológico , Imunidade Inata/efeitos dos fármacos , Imunoconjugados/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Animais , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Centro Germinativo/efeitos dos fármacos , Centro Germinativo/imunologia , Humanos , Hipertensão/imunologia , Imunidade Inata/imunologia , Imunoconjugados/genética , Imunoconjugados/imunologia , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/patologia , Camundongos , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia , Vacinas Conjugadas/administração & dosagem , Vacinas Conjugadas/efeitos adversos , Vacinas de Partículas Semelhantes a Vírus/efeitos adversos , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/imunologia
6.
Curr Opin Cardiol ; 32(6): 776-783, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28786864

RESUMO

PURPOSE OF REVIEW: Long noncoding RNAs (lncRNAs) have emerged as powerful regulators of nearly all biological processes. Their cell-type and tissue-specific expression in health and disease provides new avenues for diagnosis and therapy. This review highlights the role of lncRNAs that are involved in cardiovascular disease (CVD) with a special focus on cell types involved in cardiac injury and remodeling, vascular injury, angiogenesis, inflammation, and lipid metabolism. RECENT FINDINGS: Almost 98% of the genome does not encode for proteins. LncRNAs are among the most abundant type of RNA in the noncoding genome. Accumulating studies have uncovered novel lncRNA-mediated regulation of CVD-associated genes, signaling pathways, and pathophysiological responses. Targeting lncRNAs in vivo using short antisense oligonucleotides or by gene editing has provided important insights into disease pathogenesis through epigenetic, transcriptional, or translational mechanisms. Although cross-species conservation still remains a major obstacle, there is increasing appreciation that altered expression of lncRNAs associates with stage-specific CVD and in human patient cohorts, providing new opportunities for diagnosis and therapy. SUMMARY: A better understanding of lncRNAs will not only fundamentally improve our understanding of key signaling pathways in CVD, but also aid in the development of effective new therapies and RNA-based biomarkers.


Assuntos
Doenças Cardiovasculares/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/terapia , Células Endoteliais/metabolismo , Humanos , Metabolismo dos Lipídeos , Macrófagos/metabolismo , Monócitos/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/metabolismo
7.
Sci Rep ; 7(1): 1668, 2017 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-28490754

RESUMO

Microwave assisted with alkaline (MW-A) condition was applied in the pretreatment of swine manure, and the effect of the pretreatment on anaerobic treatment and biogas production was evaluated in this study. The two main microwaving (MW) parameters, microwaving power and reaction time, were optimized for the pretreatment. Response surface methodology (RSM) was used to investigate the effect of alkaline microwaving process for manure pretreatment at various values of pH and energy input. Results showed that the manure disintegration degree was maximized of 63.91% at energy input of 54 J/g and pH of 12.0, and variance analysis indicated that pH value played a more important role in the pretreatment than in energy input. Anaerobic digestion results demonstrated that MW-A pretreatment not only significantly increased cumulative biogas production, but also shortened the duration for a stable biogas production rate. Therefore, the alkaline microwaving pretreatment could become an alternative process for effective treatment of swine manure.


Assuntos
Álcalis/farmacologia , Biocombustíveis , Esterco/análise , Micro-Ondas , Compostos de Amônio/análise , Anaerobiose , Análise de Variância , Animais , Biodegradação Ambiental , Concentração de Íons de Hidrogênio , Metano/análise , Nitrogênio/análise , Compostos Orgânicos/análise , Solubilidade , Suínos
8.
FASEB J ; 30(9): 3216-26, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27297585

RESUMO

Thrombogenic and inflammatory mediators, such as thrombin, induce NF-κB-mediated endothelial cell (EC) activation and dysfunction, which contribute to pathogenesis of arterial thrombosis. The role of anti-inflammatory microRNA-181b (miR-181b) on thrombosis remains unknown. Our previous study demonstrated that miR-181b inhibits downstream NF-κB signaling in response to TNF-α. Here, we demonstrate that miR-181b uniquely inhibits upstream NF-κB signaling in response to thrombin. Overexpression of miR-181b inhibited thrombin-induced activation of NF-κB signaling, demonstrated by reduction of phospho-IKK-ß, -IκB-α, and p65 nuclear translocation in ECs. MiR-181b also reduced expression of NF-κB target genes VCAM-1, intercellular adhesion molecule-1, E-selectin, and tissue factor. Mechanistically, miR-181b targets caspase recruitment domain family member 10 (Card10), an adaptor protein that participates in activation of the IKK complex in response to signals transduced from protease-activated receptor-1. miR-181b reduced expression of Card10 mRNA and protein, but not protease-activated receptor-1. 3'-Untranslated region reporter assays, argonaute-2 microribonucleoprotein immunoprecipitation studies, and Card10 rescue studies revealed that Card10 is a bona fide direct miR-181b target. Small interfering RNA-mediated knockdown of Card10 expression phenocopied effects of miR-181b on NF-κB signaling and targets. Card10 deficiency did not affect TNF-α-induced activation of NF-κB signaling, which suggested stimulus-specific regulation of NF-κB signaling and endothelial responses by miR-181b in ECs. Finally, in response to photochemical injury-induced arterial thrombosis, systemic delivery of miR-181b reduced thrombus formation by 73% in carotid arteries and prolonged time to occlusion by 1.6-fold, effects recapitulated by Card10 small interfering RNA. These data demonstrate that miR-181b and Card10 are important regulators of thrombin-induced EC activation and arterial thrombosis. These studies highlight the relevance of microRNA-dependent targets in response to ligand-specific signaling in ECs.-Lin, J., He, S., Sun, X., Franck, G., Deng, Y., Yang, D., Haemmig, S., Wara, A. K. M., Icli, B., Li, D., Feinberg, M. W. MicroRNA-181b inhibits thrombin-mediated endothelial activation and arterial thrombosis by targeting caspase recruitment domain family member 10.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , MicroRNAs/metabolismo , Trombina/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Células Endoteliais , Endotélio Vascular , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Inflamação/metabolismo , Camundongos , MicroRNAs/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação , Interferência de RNA , Transdução de Sinais/fisiologia , Síndrome do Desfiladeiro Torácico , Trombina/genética , Trombose/etiologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
9.
J Hypertens ; 34(3): 474-85; discussion 485, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26771341

RESUMO

OBJECTIVE: Angiotensin II (AngII) type 1 receptor (AT1R) blockers have been proved to reduce atherosclerosis. Previously, we have invented ATRQß-001 vaccine which showed a desirable blocking effect for AT1R. The purpose of this study was to investigate whether ATRQß-001 vaccine would prevent atherosclerosis in apolipoprotein E-null (ApoE-/-) mice. METHODS: Male ApoE-/- mice were administered with ATRQß-001 vaccine, Qß virus-like particles, valsartan or vehicle over a period of 24 weeks. In vitro, human coronary artery endothelial cells preincubated with the anti-ATR-001 antibody, the neutralization antibody or valsartan for 2  h, were treated with AngII for 24  h. Histological stain and molecule biology methods were used to assess the atheroprotective effect of the vaccine. RESULTS: ATRQß-001 vaccine significantly reduced the lesion area and promoted the stability of atherosclerotic plaque. Meanwhile, macrophage infiltration as well as the expressions of adhesion molecules and monocyte chemoattractant protein-1 was obviously decreased in the ATRQß-001 vaccine group. Additionally, the vaccine markedly reduced the apoptosis in the lesions of the ApoE-/- mice. In vitro, the anti-ATR-001 antibody inhibited endothelial apoptosis induced by AngII. Furthermore, ATRQß-001 vaccine exhibited a dramatical attenuation in the expressions of lectin-like oxidized low-density lipoprotein receptor-1 and AT1R in the aortic. More importantly, compared with the valsartan group, no obvious feedback of the plasma renin-angiotensin system was elicited in the vaccine group. CONCLUSION: The results demonstrated that ATRQß-001 vaccine reduced the progression of atherosclerosis in ApoE-/- mice without obvious feedback of renin-angiotensin system.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Angiotensina II/farmacologia , Aorta/efeitos dos fármacos , Apolipoproteínas E/genética , Células Endoteliais/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/imunologia , Vacinas/farmacologia , Valsartana/farmacologia , Vasoconstritores/farmacologia , Animais , Anticorpos Neutralizantes/farmacologia , Aorta/imunologia , Aorta/patologia , Apoptose/efeitos dos fármacos , Aterosclerose/imunologia , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Quimiocina CCL2/efeitos dos fármacos , Quimiocina CCL2/imunologia , Vasos Coronários/citologia , Humanos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia , Placa Aterosclerótica/prevenção & controle , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/metabolismo , Sistema Renina-Angiotensina , Receptores Depuradores Classe E/efeitos dos fármacos , Receptores Depuradores Classe E/metabolismo
10.
J Mol Med (Berl) ; 94(2): 207-18, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26407577

RESUMO

UNLABELLED: Recently, our group has developed a therapeutic hypertensive vaccine against angiotensin (Ang) II type 1 receptor (AT1R) named ATRQß-001. To explore its potential effectiveness on streptozotocin-induced diabetic nephropathy, male Sprague Dawley rats were randomly divided into two groups: a control and a diabetic model. After 1 week, the diabetic rats were divided into four subgroups (each with 15 rats) for 14-week treatments with saline, olmesartan, ATRQß-001, and Qß virus-like particle (VLP), respectively. In addition to lower blood pressure, ATRQß-001 vaccination ameliorated biochemical parameter changes of renal dysfunction, mesangial expansion, and fibrosis through inhibiting oxidative stress, macrophage infiltration, and proinflammatory factor expression. Furthermore, ATRQß-001 vaccination suppressed renal Ang II-AT1R activation and abrogated the downregulation of angiotensin-converting enzyme 2-Ang (1-7), similar to olmesartan treatment, while no obvious feedback activation of circulating or local renin-angiotensin system (RAS) was only observed in vaccine group. In rat mesangial cells, the anti-ATR-001 antibody inhibited high glucose-induced transforming growth factor-ß1 (TGF)-ß1/Smad3 signal pathway. Additionally, no significant immune-mediated damage was detected in vaccinated animals. In conclusion, the ATRQß-001 vaccine ameliorated streptozotocin-induced diabetic renal injury via modulating two RAS axes and inhibiting TGF-ß1/Smad3 signal pathway, providing a novel, safe, and promising method to treat diabetic nephropathy. KEY MESSAGES: Overactivation of RAS plays a crucial role in the development of the DN. Our aim was to verify the effectiveness of ATRQß-001 vaccine in STZ-induced DN. The ATRQß-001 modulated two RAS axes and inhibited TGF-ß1/Smad3 signal pathway. The vaccine therapy may provide a novel, safe, and promising method to treat DN.


Assuntos
Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/prevenção & controle , Receptor Tipo 1 de Angiotensina/imunologia , Vacinas/imunologia , Angiotensina II/sangue , Animais , Biomarcadores , Análise Química do Sangue , Pressão Sanguínea , Diabetes Mellitus Experimental , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Modelos Animais de Doenças , Fibrose , Testes de Função Renal , Masculino , Fragmentos de Peptídeos/sangue , Podócitos/metabolismo , Ratos , Renina/sangue , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/metabolismo , Estreptozocina/efeitos adversos , Fator de Crescimento Transformador beta1/metabolismo , Vacinas/administração & dosagem
11.
J Hypertens ; 32(4): 762-70, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24609215

RESUMO

BACKGROUND: Endothelial microparticles (EMPs) are small vesicular structures that serve as a marker of endothelial function. Angiotensin II receptor type 1 autoantibody (AT1-AA) can cause endothelial dysfunction. However, whether AT1-AA promotes EMPs formation and the mechanism remains obscure. METHODS: The titres of sera AT1-AA of 126 hypertensive patients and 30 normotensive individuals were evaluated by ELISA. EMPs in the sera and the supernatants of human umbilical vein endothelial cells (HUVECs) were measured by flow cytometry. The phosphorylation levels of mitogen-activated protein kinase (MAPK) pathways in HUVECs treated by AT1-AA were assessed and their correlation with microparticle formation was also analysed. Furthermore, the production of intracellular reactive oxygen species (ROS) and nitric oxide in HUVECs was examined after incubation with 'injured' endothelial microparticle (iEMPs) (EMPs derived from AT1-AA treated HUVECs). RESULTS: The positive rate of AT1-AA in 126 hypertensive patients was 21.4% (27/126), and higher than that in normotensive individuals [3.3% (1/30), P < 0.01]. Circulating EMP (CD31+/CD42b-) levels were corresponding to the AT1-AA titres in hypertensive group (r(2) = 0.3661, P < 0.01). AT1-AA promoted EMPs generation from HUVECs in a time and dose-dependent manner than the vehicle or nonspecific IgG. Meanwhile, AT1-AA significantly elevated phosphorylation level of P38 and ERK in HUVECs. Lorsartan and P38 inhibitor could suppress the AT1-AA's stimulation effect on EMPs generation. Moreover, the iEMP greatly increased ROS production and reduced nitric oxide synthesis in HUVECs. CONCLUSION: Our findings showed that circulating EMPs levels positively correlate to the serum AT1-AA titres in essential hypertension patients. The AT1-AA could promote EMPs generation in HUVECs through activation of P38 MAPK signalling pathway, and this effect could be effectively inhibited by losartan or p38 inhibitor. Angiotensin receptor blockers (ARBs) may be more suitable for AT1-AA(+) hypertensive patients on account of suppressing the AT1-AA's stimulation effect on EMPs generation.


Assuntos
Autoanticorpos/metabolismo , Regulação da Expressão Gênica , Hipertensão/sangue , Receptor Tipo 1 de Angiotensina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Adulto , Autoanticorpos/sangue , Células Endoteliais/citologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Imunoglobulina G/metabolismo , Masculino , Pessoa de Meia-Idade , Óxido Nítrico/metabolismo , Fosforilação , Espécies Reativas de Oxigênio , Receptor Tipo 1 de Angiotensina/imunologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...