Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Circulation ; 146(18): 1367-1382, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36172862

RESUMO

BACKGROUND: Hypertension is a common cardiovascular disease that is related to genetic and environmental factors, but its mechanisms remain unclear. DNA methylation, a classic epigenetic modification, not only regulates gene expression but is also susceptible to environmental factors, linking environmental factors to genetic modification. Therefore, globally screening differential genomic DNA methylation in patients with hypertension is important for investigating hypertension mechanisms. METHODS: Differential genomic DNA methylation in patients with hypertension, individuals with prehypertension, and healthy control individuals was screened using Illumina 450K BeadChip and verified by pyrosequencing. Plasma OVGP1 (oviduct glycoprotein 1) levels were determined using an enzyme-linked immunosorbent assay. Ovgp1 transgenic and knockout mice were generated to analyze the function of OVGP1. The blood pressure levels of the mouse models were measured using the tail-cuff system and radiotelemetry methods. The role of OVGP1 in vascular remodeling was determined by vascular relaxation studies. Protein-protein interactions were investigated using a pull-down/mass spectrometry assay and verified with coimmunoprecipitation and pull-down assays. RESULTS: We found a hypomethylated site at cg20823859 in the promoter region of OVGP1 and plasma OVGP1 levels were significantly increased in patients with hypertension. This finding indicates that OVGP1 is associated with hypertension. In Ovgp1 transgenic mice, OVGP1 overexpression caused an increase in blood pressure, dysfunctional vasoconstriction and vasodilation, remodeling of arterial walls, and increased vascular superoxide stress and inflammation, and these phenomena were exacerbated by angiotensin II infusion. In contrast, OVGP1 deficiency attenuated angiotensin II-induced vascular oxidase stress, inflammation, and collagen deposition. These findings indicate that OVGP1 is a prohypertensive factor that directly promotes vascular remodeling. Pull-down and coimmunoprecipitation assays showed that MYH9 (nonmuscle myosin heavy chain IIA) interacted with OVGP1, whereas inhibition of MYH9 attenuated OVGP1-induced hypertension and vascular remodeling. CONCLUSIONS: Hypomethylation at cg20823859 in the promoter region of OVGP1 is associated with hypertension and induces upregulation of OVGP1. The interaction between OVGP1 and MYH9 contributes to vascular remodeling and dysfunction. Therefore, OVGP1 is a prohypertensive factor that promotes vascular remodeling by binding with MYH9.


Assuntos
Hipertensão , Remodelação Vascular , Camundongos , Animais , Angiotensina II/farmacologia , Proteínas do Citoesqueleto , Camundongos Knockout , Camundongos Transgênicos , Glicoproteínas/genética , Inflamação , Cadeias Pesadas de Miosina/genética
2.
Front Neurosci ; 16: 1002590, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36523430

RESUMO

Background: The roles and potential diagnostic value of circRNAs in intracerebral hemorrhage (ICH) remain elusive. Methods: This study aims to investigate the expression profiles of circRNAs by RNA sequencing and RT-PCR in a discovery cohort and an independent validation cohort. Bioinformatics analysis was performed to identify the potential functions of circRNA host genes. Machine learning classification models were used to assess circRNAs as potential biomarkers of ICH. Results: A total of 125 and 284 differentially expressed circRNAs (fold change > 1.5 and FDR < 0.05) were found between ICH patients and healthy controls in the discovery and validation cohorts, respectively. Nine circRNAs were consistently altered in ICH patients compared to healthy controls. The combination of the novel circERBB2 and circCHST12 in ICH patients and healthy controls showed an area under the curve of 0.917 (95% CI: 0.869-0.965), with a sensitivity of 87.5% and a specificity of 82%. In combination with ICH risk factors, circRNAs improved the performance in discriminating ICH patients from healthy controls. Together with hsa_circ_0005505, two novel circRNAs for differentiating between patients with ICH and healthy controls showed an AUC of 0.946 (95% CI: 0.910-0.982), with a sensitivity of 89.1% and a specificity of 86%. Conclusion: We provided a transcriptome-wide overview of aberrantly expressed circRNAs in ICH patients and identified hsa_circ_0005505 and novel circERBB2 and circCHST12 as potential biomarkers for diagnosing ICH.

3.
Theranostics ; 11(19): 9587-9604, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34646388

RESUMO

Rationale: The progressive disruption of extracellular matrix (ECM) proteins, particularly early elastin fragmentation followed by abnormalities in collagen fibril organization, are key pathological processes that contribute to dissecting abdominal aortic aneurysm (AAA) pathogenesis. Lysyl hydroxylase 1 (LH1) is essential for type I/III collagen intermolecular crosslinking and stabilization. However, its function in dissecting AAA has not been explored. Here, we investigated whether LH1 is significantly implicated in dissecting AAA progression and therapeutic intervention. Methods and Results: Sixteen-week-old male LH1-deficient and wild-type (WT) mice on the C57Bl/6NCrl background were infused with angiotensin II (Ang II, 1000 ng/kg per minute) via subcutaneously implanted osmotic pumps for 4 weeks. Ang II increased LH1 levels in the abdominal aortas of WT mice, whereas mice lacking LH1 developed dissecting AAA. To evaluate the related mechanism, we performed whole-transcriptomic analysis, which demonstrated that LH1 deficiency aggravated gene transcription alterations; in particular, the expression of thrombospondin-1 was markedly upregulated in the aortas of LH1-deficient mice. Furthermore, targeting thrombospondin-1 with TAX2 strongly inhibited the proinflammatory process, matrix metalloproteinase (MMP) activity and vascular smooth muscle cells (VSMCs) apoptosis, ultimately decreasing the incidence of dissecting AAA. Restoration of LH1 protein expression in LH1-deficient mice by intraperitoneal injection of an adeno-associated virus normalized thrombospondin-1 levels, subsequently alleviating dissecting AAA formation and preserving aortic structure and function. Consistently, in human AAA specimens, decreased LH1 expression was associated with increased thrombospondin-1 levels. Conclusions: LH1 deficiency contributes to dissecting AAA pathogenesis, at least in part, by upregulating thrombospondin-1 expression, which subsequently enables proinflammatory processes, MMP activation and VSMCs apoptosis. Our study provides evidence that LH1 is a potential critical therapeutic target for AAA.


Assuntos
Aneurisma da Aorta Abdominal/metabolismo , Dissecção Aórtica/metabolismo , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Angiotensina II/farmacologia , Animais , Aorta/metabolismo , Apolipoproteínas E/genética , Apoptose/efeitos dos fármacos , Colágeno/metabolismo , Modelos Animais de Doenças , Matriz Extracelular/fisiologia , Expressão Gênica/genética , Inflamação/metabolismo , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/metabolismo , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/deficiência , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/genética , Trombospondina 1/genética , Trombospondina 1/metabolismo , Transcriptoma/genética
4.
Front Physiol ; 12: 770223, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34867478

RESUMO

Background: We used a targeted metabolomics approach to identify fatty acid (FA) metabolites that distinguished patients with coronary artery ectasia (CAE) from healthy Controls and patients with coronary artery disease (CAD). Materials and methods: Two hundred fifty-two human subjects were enrolled in our study, such as patients with CAE, patients with CAD, and Controls. All the subjects were diagnosed by coronary angiography. Plasma metabolomic profiles of FAs were determined by an ultra-high-performance liquid chromatography coupled to triple quadrupole mass spectrometric (UPLC-QqQ-MS/MS). Results: Ninety-nine plasma metabolites were profiled in the discovery sets (n = 72), such as 35 metabolites of arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), 10 FAs, and 54 phospholipids. Among these metabolites, 36 metabolites of AA, EPA, and DHA showed the largest difference between CAE and Controls or CAD. 12-hydroxyeicosatetraenoic acid (12-HETE), 17(S)-hydroxydocosahexaenoic acid (17-HDoHE), EPA, AA, and 5-HETE were defined as a biomarker panel in peripheral blood to distinguish CAE from CAD and Controls in a discovery set (n = 72) and a validation set (n = 180). This biomarker panel had a better diagnostic performance than metabolite alone in differentiating CAE from Controls and CAD. The areas under the ROC curve of the biomarker panel were 0.991 and 0.836 for CAE versus Controls and 1.00 and 0.904 for CAE versus CAD in the discovery and validation sets, respectively. Conclusions: Our findings revealed that the metabolic profiles of FAs in the plasma from patients with CAE can be distinguished from those of Controls and CAD. Differences in FAs metabolites may help to interpret pathological mechanisms of CAE.

5.
Epigenomics ; 13(5): 379-395, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33507103

RESUMO

Aim: To investigate the expression profiles of circRNAs after intracerebral hemorrhage (ICH). Materials & methods: RNA sequencing and qRT-PCR were used to investigate and validate circRNA expression levels. Bioinformatics analysis was performed to explore potential functions of the circRNAs. Results: Expression levels of 15 circRNAs were consistently altered in patients with ICH compared with their expression levels in hypertension. Three circRNAs, hsa_circ_0001240, hsa_circ_0001947 and hsa_circ_0001386, individually or combined, were confirmed as promising biomarkers for predicting and diagnosing ICH. The circRNAs were involved mainly in lysine degradation and the immune system. Conclusion: This is the first study to report expression profiles of circRNAs after ICH and to propose that three circRNAs are potential biomarkers for ICH.


Assuntos
Hemorragia Cerebral/genética , Hipertensão/genética , RNA Circular/sangue , Biomarcadores/sangue , Hemorragia Cerebral/sangue , Feminino , Humanos , Hipertensão/sangue , Integrinas/metabolismo , Lisina/metabolismo , Masculino , MicroRNAs , Pessoa de Meia-Idade , RNA Mensageiro
6.
Hypertension ; 75(3): 680-692, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31928110

RESUMO

Plasma metabolic profiles were compared between patients with hypertension with and without left ventricular hypertrophy and significantly decreased oleic acid (OA) levels were observed in the peripheral blood of patients with hypertension with left ventricular hypertrophy. We sought to determine the effect and underlying mechanisms of OA on cardiac remodeling. In vitro studies with isolated neonatal mouse cardiomyocytes and cardiac fibroblasts revealed that OA significantly attenuated Ang II (angiotensin II)-induced cardiomyocyte growth and cardiac fibroblast collagen expression. In vivo, cardiac function, hypertrophic growth of cardiomyocytes, and fibrosis were analyzed after an Ang II (1000 ng/kg/minute) pump was implanted for 14 days. We found that OA could significantly prevent Ang II-induced cardiac remodeling in mice. RNA sequencing served as a gene expression roadmap highlighting gene expression changes in the hearts of Ang II-induced mice and OA-treated mice. The results revealed that FGF23 (fibroblast growth factor 23) expression was significantly upregulated in mouse hearts in response to Ang II infusion, which was significantly suppressed in the hearts of OA-treated mice. Furthermore, overexpression of FGF23 in the heart by injection of an AAV-9 vector aggravated Ang II-induced cardiac remodeling and impaired the protective effect of OA on cardiac remodeling. Further study found that OA could suppress Ang II-induced FGF23 expression by inhibiting the translocation of Nurr1 (nuclear receptor-related 1 protein) from the cytoplasm to the nucleus. Our findings suggest a novel role of OA in preventing Ang II-induced cardiac remodeling via suppression of FGF23 expression.


Assuntos
Angiotensina II/farmacologia , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Hipertrofia Ventricular Esquerda/sangue , Ácido Oleico/fisiologia , Remodelação Ventricular/fisiologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Colágeno/biossíntese , Dependovirus/genética , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/biossíntese , Fatores de Crescimento de Fibroblastos/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Ontologia Genética , Vetores Genéticos , Humanos , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Ácido Oleico/sangue , Ácido Oleico/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Organismos Livres de Patógenos Específicos , Remodelação Ventricular/efeitos dos fármacos
7.
J Cardiovasc Transl Res ; 13(5): 783-789, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31828536

RESUMO

The study aimed to investigate the relationship between mtDNA copy number and the risk of all-cause mortality in stroke. One thousand four hundred eighty-four stroke patients were documented including 273 deaths (127 thrombosis, 52 lacunar, 94 hemorrhage). Patients in the third quartile had the lowest mortality rates in overall stroke and the three subtypes. The lowest quartile of mtDNA copy number (Q1 < 85.85) indicated an increased risk of all-cause mortality in stroke patients (adjusted HR, 1.52; 95% CI, 1.08-2.14; p = 0.017). In the subtype analysis, the risk of all-cause mortality appeared only in lacunar infarct, and the patients in the Q1 (< 87.76) and Q4 (> 150.61) mtDNA copy number groups showed significantly higher risks of HRs (Q1, adjusted HR, 3.87, 95% CI, 1.52-9.83; Q4, adjusted HR, 3.08, 95% CI, 1.16-8.18). Stroke patients with lacunar infarct in mtDNA copy number < 87.76 or > 150.61 were at a high risk of poor outcomes in all-cause mortality.


Assuntos
Isquemia Encefálica/genética , Hemorragia Cerebral/genética , DNA Mitocondrial/genética , Dosagem de Genes , Trombose Intracraniana/genética , Acidente Vascular Cerebral Lacunar/genética , Idoso , Isquemia Encefálica/diagnóstico por imagem , Isquemia Encefálica/mortalidade , Hemorragia Cerebral/diagnóstico por imagem , Hemorragia Cerebral/mortalidade , China , DNA Mitocondrial/sangue , Feminino , Humanos , Trombose Intracraniana/diagnóstico por imagem , Trombose Intracraniana/mortalidade , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Acidente Vascular Cerebral Lacunar/diagnóstico por imagem , Acidente Vascular Cerebral Lacunar/mortalidade
8.
Transl Stroke Res ; 11(5): 1077-1094, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32323149

RESUMO

Intracerebral hemorrhage (ICH) is a catastrophic stroke with high mortality, and the mechanism underlying ICH is largely unknown. Previous studies have shown that high serum uric acid (SUA) levels are an independent risk factor for hypertension, cardiovascular disease (CVD), and ischemic stroke. However, our metabolomics data showed that SUA levels were lower in recurrent intracerebral hemorrhage (R-ICH) patients than in ICH patients, indicating that lower SUA might contribute to ICH. In this study, we confirmed the association between low SUA levels and the risk for recurrence of ICH and for cardiac-cerebral vascular mortality in hypertensive patients. To determine the mechanism by which low SUA effects ICH pathogenesis, we developed the first low SUA mouse model and conducted transcriptome profiling of the cerebrovasculature of ICH mice. When combining these assessments with pathological morphology, we found that low SUA levels led to ICH in mice with angiotensin II (Ang II)-induced hypertension and aggravated the pathological progression of ICH. In vitro, our results showed that p-Erk1/2-MMP axis were involved in the low UA-induce degradation of elastin, and that physiological concentrations of UA and p-Erk1/2-specific inhibitor exerted a protective role. This is the first report describing to the disruption of the smooth muscle cell (SMC)-elastin contractile units in ICH. Most importantly, we revealed that the upregulation of the p-Erk1/2-MMP axis, which promotes the degradation of elastin, plays a vital role in mediating low SUA levels to exacerbate cerebrovascular rupture during the ICH process.


Assuntos
Hemorragia Cerebral/sangue , Hemorragia Intracraniana Hipertensiva/sangue , Miócitos de Músculo Liso/metabolismo , Acidente Vascular Cerebral/sangue , Ácido Úrico/sangue , Animais , Hemorragia Cerebral/patologia , Humanos , Hipertensão/sangue , Sistema de Sinalização das MAP Quinases/fisiologia , Metaloproteinases da Matriz/metabolismo , Camundongos , Fatores de Risco , Acidente Vascular Cerebral/patologia , Regulação para Cima
9.
Nat Commun ; 11(1): 3218, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32587239

RESUMO

Unruptured intracranial aneurysm (UIA) is a life-threatening cerebrovascular condition. Whether changes in gut microbial composition participate in the development of UIAs remains largely unknown. We perform a case-control metagenome-wide association study in two cohorts of Chinese UIA patients and control individuals and mice that receive fecal transplants from human donors. After fecal transplantation, the UIA microbiota is sufficient to induce UIAs in mice. We identify UIA-associated gut microbial species link to changes in circulating taurine. Specifically, the abundance of Hungatella hathewayi is markedly decreased and positively correlated with the circulating taurine concentration in both humans and mice. Consistently, gavage with H. hathewayi normalizes the taurine levels in serum and protects mice against the formation and rupture of intracranial aneurysms. Taurine supplementation also reverses the progression of intracranial aneurysms. Our findings provide insights into a potential role of H. hathewayi-associated taurine depletion as a key factor in the pathogenesis of UIAs.


Assuntos
Clostridiaceae/metabolismo , Microbioma Gastrointestinal , Aneurisma Intracraniano , Taurina/metabolismo , Animais , Estudos de Casos e Controles , Estudos de Coortes , Progressão da Doença , Transplante de Microbiota Fecal , Feminino , Humanos , Aneurisma Intracraniano/microbiologia , Aneurisma Intracraniano/patologia , Masculino , Camundongos , Prognóstico , Fatores de Risco
10.
J Cereb Blood Flow Metab ; 39(3): 547-561, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30516406

RESUMO

Hypertensive intracerebral hemorrhage (ICH) is a devastating cerebrovascular disease with no effective treatment. Lysyl hydroxylase 3 (LH3) is essential for collagen IV intermolecular crosslinking and stabilization. Deficiency in LH3 affects the assembly and secretion of collagen IV and basement membrane (BM) integrity of vessels. Here, we investigated whether LH3 has significant implications for disease progression and therapeutic intervention. Spontaneous hypertensive ICH of mice was induced by angiotensin II and L-NAME treatment. The adeno-associated virus was delivered into brain by stereotactic injection to knockdown or overexpress LH3. We found LH3 levels were reduced in human patients with ICH and gradually decreased in mice before ICH. LH3 knockdown increased the incidence of hypertensive ICH in mice. The incidence, number, and size of ICHs in mice were markedly reduced by LH3 overexpression. RNA-seq revealed that LH3 overexpression significantly reversed the profound alterations in gene transcriptional profiles of cerebral vessels. LH3 overexpression was sufficient to enhance BM integrity, inhibit matrix metalloproteinase activity, attenuate microglial activation and leukocyte infiltration, and reduce VSMC apoptosis before ICH. These results indicate that LH3 overexpression attenuates susceptibility to hypertensive ICH. We emphasize that LH3 modulation may serve as a viable approach for future investigations of ICH prevention.


Assuntos
Hemorragia Intracraniana Hipertensiva/prevenção & controle , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Animais , Membrana Basal/irrigação sanguínea , Vasos Sanguíneos/efeitos dos fármacos , Progressão da Doença , Suscetibilidade a Doenças , Humanos , Incidência , Hemorragia Intracraniana Hipertensiva/induzido quimicamente , Hemorragia Intracraniana Hipertensiva/tratamento farmacológico , Camundongos , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/administração & dosagem , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/farmacologia
11.
DNA Cell Biol ; 37(5): 426-431, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29461087

RESUMO

Vascular remodeling is a key process leading to arterial stenosis. Ufmylation, a novel ubiquitin-like modification, was observed to be associated with many biological processes. However, whether ufmylation is involved in the regulation of vascular remodeling remains unclear. Therefore, the present study focused on the role of ufmylation in vascular remodeling. Mouse femoral artery guidewire injury models were used for inducing vascular remodeling. We found that the expression of Ufm1 was upregulated in hyperplastic neointima. By treating vascular smooth muscle cells (VSMCs) with platelet-derived growth factor BB (PDGF-BB) for 3, 6, 12, and 24 h, respectively, we observed that ufmylation was significantly activated in a time-dependent manner. Consistently, the expression levels of Ufc1, Ufl1, and Ufbp1, as key components of the ufmylation system, were all upregulated by PDGF-BB. In contrast, knockdown of Ufm1 expression attenuated PDGF-BB-induced VSMC proliferation. In addition, we observed that ufmylation was activated by lipopolysaccharide (LPS) in endothelial cells, whereas knockdown of Ufm1 was synergized with LPS-induced endothelial cell injury. These findings indicate that ufmylation may participate in regulation of the VSMC phenotypic switch and endothelial cell injury, which may help in the understanding of vascular remodeling.


Assuntos
Células Endoteliais/metabolismo , Proteínas/metabolismo , Remodelação Vascular , Vasculite/metabolismo , Animais , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Processamento de Proteína Pós-Traducional , Proteínas/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Remodelação Vascular/efeitos dos fármacos , Remodelação Vascular/genética , Vasculite/induzido quimicamente , Vasculite/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA