Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Gene Med ; 26(1): e3656, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38282147

RESUMEN

BACKGROUND: The induction of cardiomyocyte (CM) proliferation is a promising approach for cardiac regeneration following myocardial injury. MicroRNAs (miRNAs) have been reported to regulate CM proliferation. In particular, miR-431 expression decreases during cardiac development, according to Gene Expression Omnibus (GEO) microarray data. However, whether miR-431 regulates CM proliferation has not been thoroughly investigated. METHODS: We used integrated bioinformatics analysis of GEO datasets to identify the most significantly differentially expressed miRNAs. Real-time quantitative PCR and fluorescence in situ hybridization were performed to determine the miRNA expression patterns in hearts. Gain- and loss-of-function assays were conducted to detect the role of miRNA in CM proliferation. Additionally, we detected whether miR-431 affected CM proliferation in a myocardial infarction model. The TargetScan, miRDB and miRWalk online databases were used to predict the potential target genes of miRNAs. Luciferase reporter assays were used to study miRNA interactions with the targeting mRNA. RESULTS: First, we found a significant reduction in miR-431 levels during cardiac development. Then, by overexpression and inhibition of miR-431, we demonstrated that miR-431 promotes CM proliferation in vitro and in vivo, as determined by immunofluorescence assays of 5-ethynyl-2'-deoxyuridine (EdU), pH3, Aurora B and CM count, whereas miR-431 inhibition suppresses CM proliferation. Then, we found that miR-431 improved cardiac function post-myocardial infarction. In addition, we identified FBXO32 as a direct target gene of miR-431, with FBXO32 mRNA and protein expression being suppressed by miR-431. FBXO32 inhibited CM proliferation. Overexpression of FBXO32 blocks the enhanced effect of miR-431 on CM proliferation, suggesting that FBXO32 is a functional target of miR-431 during CM proliferation. CONCLUSION: In summary, miR-431 promotes CM proliferation by targeting FBXO32, providing a potential molecular target for preventing myocardial injury.


Asunto(s)
MicroARNs , Proteínas Musculares , Infarto del Miocardio , Miocitos Cardíacos , Proteínas Ligasas SKP Cullina F-box , Proliferación Celular/genética , Hibridación Fluorescente in Situ , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Musculares/genética , Infarto del Miocardio/genética , Miocitos Cardíacos/citología , ARN Mensajero/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Animales
2.
Mol Ther ; 30(11): 3477-3498, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-35791879

RESUMEN

Reactive oxygen species (ROS) derived from oxygen-dependent mitochondrial metabolism are the essential drivers of cardiomyocyte (CM) cell-cycle arrest in adulthood. Mitochondria-localized circular RNAs (circRNAs) play important roles in regulating mitochondria-derived ROS production, but their functions in cardiac regeneration are still unknown. Herein, we investigated the functions and underlying mechanism of mitochondria-localized circSamd4 in cardiac regeneration. We found that circSamd4 was selectively expressed in fetal and neonatal CMs. The transcription factor Nrf2 controlled circSamd4 expression by binding to the promoter of circSamd4 host gene. CircSamd4 overexpression reduced while circSamd4 silenced increased mitochondrial oxidative stress and subsequent oxidative DNA damage. Moreover, circSamd4 overexpression induced CM proliferation and prevented CM apoptosis, which reduced the size of the fibrotic area and improved cardiac function after myocardial infarction (MI). Mechanistically, circSamd4 reduced oxidative stress generation and maintained mitochondrial dynamics by inducing the mitochondrial translocation of the Vcp protein, which downregulated Vdac1 expression and prevented the mitochondrial permeability transition pore (mPTP) from opening. Our findings suggest that circSamd4 is a novel therapeutic target for heart failure after MI.


Asunto(s)
Infarto del Miocardio , ARN Circular , Humanos , Recién Nacido , Adulto , ARN Circular/genética , Especies Reactivas de Oxígeno/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Miocitos Cardíacos/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/terapia , Infarto del Miocardio/metabolismo
3.
BMC Nephrol ; 23(1): 32, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-35033017

RESUMEN

BACKGROUND: To verify that the single nucleotide polymorphisms (SNP) of vitamin D receptor (VDR) may lead to genetic susceptibility to left ventricular hypertrophy (LVH), the present study was designed to study four SNPs of VDR associated with LVH in maintenance hemodialysis (MHD) patients of Han nationality. METHODS: 120 MHD patients were recruited at Department of Nephrology, Zhongnan Hospital of Wuhan University to analyze the expression of genotype, allele and haplotype of Fok I, Bsm I, Apa I and Taq I in blood samples, and to explore their correlation with blood biochemical indexes and ventricular remodeling. RESULTS: The results showed that the risks of CVD included gender, dialysis time, heart rate, SBP, glycated hemoglobin, calcium, iPTH and CRP concentration. Moreover, LAD, LVDd, LVDs, IVST and LVMI in B allele of Bsm I increased significantly. Fok I, Apa I and Taq I polymorphisms have no significant difference between MHD with LVH and without LVH. Further study showed that VDR expression level decreased significantly in MHD patients with LVH, and the B allele was positively correlated with VDR Expression. CONCLUSION: VDR Bsm I gene polymorphism may predict cardiovascular disease risk of MDH patients, and provided theoretical basis for early detection and prevention of cardiovascular complications.


Asunto(s)
Hipertrofia Ventricular Izquierda/genética , Polimorfismo de Nucleótido Simple , Receptores de Calcitriol/genética , Diálisis Renal , Adulto , China , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico
4.
Circulation ; 139(25): 2857-2876, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-30947518

RESUMEN

BACKGROUND: circRNAs (circular RNAs) are emerging as powerful regulators of cardiac development and disease, but their roles in cardiac regeneration are still unknown. This study used superenhancers to distinguish key circRNAs in the regulation of cardiac regeneration and explored the mechanisms underlying circRNA functions. METHODS: We used integrated bioinformatics analysis of RNA sequencing data and superenhancer catalogs to identify superenhancer-associated circRNAs. Quantitative polymerase chain reactions and in situ hybridization were performed to determine the circRNA expression patterns in hearts. Gain- and loss-of-function assays were conducted to detect the role of circRNAs in cardiomyocyte proliferation and cardiac repair after myocardial infarction. Chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assays were used to determine the binding of Meis1 (Meis homeobox 1) on circNfix-associated superenhancers. RNA pulldown and luciferase reporter assays were used to study circRNA interactions with proteins and miRNAs (micro RNAs). RESULTS: We identified a circRNA, Nfix circRNA (circNfix), that was regulated by a superenhancer and overexpressed in the adult heart in humans, rats, and mice. The transcription factor Meis1 bound to the superenhancer at the circNfix locus, and increased its expression. In vitro and in vivo, cardiomyocyte proliferation was increased by knockdown of circNfix, whereas it was inhibited by circNfix overexpression. Moreover, circNfix downregulation promoted cardiomyocyte proliferation and angiogenesis and inhibited cardiomyocyte apoptosis after myocardial infarction, attenuating cardiac dysfunction and improving the prognosis. Mechanistically, circNfix reinforced the interaction of Ybx1 (Y-box binding protein 1) with Nedd4l (an E3 ubiquitin ligase), and induced Ybx1 degradation through ubiquitination, repressing cyclin A2 and cyclin B1 expression. In addition, circNfix acted as a sponge for miR-214 to promote Gsk3ß (glycogen synthase kinase 3 ß) expression and repress ß-catenin activity. CONCLUSIONS: Loss of superenhancer-regulated circNfix promotes cardiac regenerative repair and functional recovery after myocardial infarction by suppressing Ybx1 ubiquitin-dependent degradation and increasing miR-214 activity and thus may be a promising strategy for improving the prognosis after MI.


Asunto(s)
Proliferación Celular , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , ARN Circular/metabolismo , Regeneración , Animales , Apoptosis , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Ubiquitina-Proteína Ligasas Nedd4/genética , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Neovascularización Fisiológica , ARN Circular/genética , Ratas Sprague-Dawley , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 39(1): e10-e25, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30580562

RESUMEN

Objective- Vascular smooth muscle cell phenotypic transition plays a critical role in the formation of abdominal aortic aneurysms (AAAs). SM22α (smooth muscle 22α) has a vital role in maintaining the smooth muscle cell phenotype and is downregulated in AAA. However, whether manipulation of the SM22α gene influences the pathogenesis of AAA is unclear. Here, we investigated whether SM22α prevents AAA formation and explored the underlying mechanisms. Approach and Results- In both human and animal AAA tissues, a smooth muscle cell phenotypic switch was confirmed, as manifested by the downregulation of SM22α and α-SMA (α-smooth muscle actin) proteins. The methylation level of the SM22α gene promoter was dramatically higher in mouse AAA tissues than in control tissues. SM22α knockdown in ApoE-/- (apolipoprotein E-deficient) mice treated with Ang II (angiotensin II) accelerated the formation of AAAs, as evidenced by a larger maximal aortic diameter and more medial elastin degradation than those found in control mice, whereas SM22α overexpression exerted opposite effects. Similar results were obtained in a calcium chloride-induced mouse AAA model. Mechanistically, SM22α deficiency significantly increased reactive oxygen species production and NF-κB (nuclear factor-κB) activation in AAA tissues, whereas SM22α overexpression produced opposite effects. NF-κB antagonist SN50 or antioxidant N-acetyl-L-cysteine partially abrogated the exacerbating effects of SM22α silencing on AAA formation. Conclusions- SM22α reduction in AAAs because of the SM22α promoter hypermethylation accelerates AAA formation through the reactive oxygen species/NF-κB pathway, and therapeutic approaches to increase SM22α expression are potentially beneficial for preventing AAA formation.


Asunto(s)
Aneurisma de la Aorta Abdominal/prevención & control , Proteínas de Microfilamentos/fisiología , Proteínas Musculares/fisiología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , FN-kappa B/fisiología , Especies Reactivas de Oxígeno/metabolismo , Animales , Células Cultivadas , Metilación de ADN , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , NADPH Oxidasas/metabolismo , Fenotipo , Fosforilación , Regiones Promotoras Genéticas
6.
Mol Ther ; 27(1): 29-45, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30528086

RESUMEN

Reactivating post-natal myocardial regeneration potential may be a feasible strategy to regenerate the injured adult heart. Long non-coding RNAs (lncRNAs) have been implicated in regulating cellular differentiation, but whether they can elicit a regenerative response in the post-natal heart remains unknown. In this study, by characterizing the lncRNA transcriptome in human hearts during the fetal-to-adult transition, we found that 3,092 lncRNAs were differentially expressed, and we further identified a novel upregulated fetal lncRNA that we called endogenous cardiac regeneration-associated regulator (ECRAR), which promoted DNA synthesis, mitosis, and cytokinesis in post-natal day 7 and adult rat cardiomyocytes (CMs). Overexpression of ECRAR markedly stimulated myocardial regeneration and induced recovery of cardiac function after myocardial infarction (MI). Knockdown of ECRAR inhibited post-natal day 1 CM proliferation and prevented post-MI recovery. ECRAR was transcriptionally upregulated by E2F transcription factor 1 (E2F1). In addition, ECRAR directly bound to and promoted the phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2), resulting in downstream targets of cyclin D1 and cyclin E1 activation, which, in turn, activated E2F1. The E2F1-ECRAR-ERK1/2 signaling formed a positive feedback loop to drive cell cycle progression, and, therefore, it promoted CM proliferation. These findings indicated that our newly discovered ECRAR may be a valuable therapeutic target for heart failure.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Miocardio/citología , Miocardio/metabolismo , ARN Largo no Codificante/metabolismo , Regeneración/fisiología , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Corazón Fetal/citología , Corazón Fetal/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , Ratas , Ratas Wistar , Regeneración/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
7.
Clin Sci (Lond) ; 133(13): 1439-1455, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31235554

RESUMEN

The long non-coding RNA (lncRNA) PTENP1 is a pseudogene of phosphatase and tensin homologue deleted on chromosome ten (PTEN), has been implicated in smooth muscle cell (SMC) proliferation and apoptosis. PTENP1 is the pseudogene of PTEN. However, it is unclear whether and how PTENP1 functions in the proliferation and apoptosis of human aortic SMCs (HASMCs). Here, we hypothesised that PTENP1 inhibits HASMC proliferation and enhances apoptosis by promoting PTEN expression. PCR analysis and Western blot assays respectively showed that both PTENP1 and PTEN were up-regulated in human aortic dissection (AD) samples. PTENP1 overexpression significantly increased the protein expression of PTEN, promoted apoptosis and inhibited the proliferation of HASMCs. PTENP1 silencing exhibited the opposite effects and mitigated H2O2-induced apoptosis of HASMCs. In an angiotensin II (Ang II)-induced mouse aortic aneurysm (AA) model, PTENP1 overexpression potentiated aortic SMC apoptosis, exacerbated aneurysm formation. Mechanistically, RNA pull-down assay and a series of luciferase reporter assays using miR-21 mimics or inhibitors identified PTENP1 as a molecular sponge for miR-21 to endogenously compete for the binding between miR-21 and the PTEN transcript, releasing PTEN expression. This finding was further supported by in vitro immunofluorescent evidence showing decreased cell apoptosis upon miR-21 mimic administration under baseline PTENP1 overexpression. Ex vivo rescue of PTEN significantly mitigated the SMC apoptosis induced by PTENP1 overexpression. Finally, Western blot assays showed substantially reduced Akt phosphorylation and cyclin D1 and cyclin E levels with up-regulated PTENP1 in HASMCs. Our study identified PTENP1 as a mediator of HASMC homeostasis and suggests that PTENP1 is a potential target in AD or AA intervention.


Asunto(s)
Aneurisma de la Aorta/metabolismo , Disección Aórtica/metabolismo , Apoptosis , Proliferación Celular , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Seudogenes , ARN Largo no Codificante/metabolismo , Disección Aórtica/genética , Disección Aórtica/patología , Animales , Aorta/metabolismo , Aorta/patología , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/patología , Ciclo Celular , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal
8.
Crit Care Med ; 46(9): e912-e920, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29965834

RESUMEN

OBJECTIVES: Effective treatment for microvascular thrombosis-induced coronary no-reflow remains an unmet clinical need. This study sought to evaluate whether diagnostic ultrasound and microbubbles treatment could improve outcomes of coronary no-reflow by dissolving platelet- and erythrocyte-rich microthrombi. DESIGN: Randomized controlled laboratory investigation. SETTING: Research laboratory. SUBJECTS: Mongrel dogs. INTERVENTIONS: Coronary no-reflow models induced by platelet- or erythrocyte-rich microthrombi were established and randomly assigned to control, ultrasound, recombinant tissue-type plasminogen activator, ultrasound + microbubbles, or ultrasound + microbubbles + recombinant tissue-type plasminogen activator group. All treatments lasted for 30 minutes. MEASUREMENTS AND MAIN RESULTS: Percentage of microemboli-obstructed coronary arterioles was lower in ultrasound + microbubbles group than that in control group for platelet- (> 50% obstruction: 10.20% ± 3.56% vs 31.80% ± 3.96%; < 50% obstruction: 14.80% ± 4.15% vs 28.20% ± 3.56%) and erythrocyte-rich microthrombi (> 50% obstruction: 8.20% ± 3.11% vs 30.60% ± 4.83%; < 50% obstruction: 12.80% ± 4.15% vs 25.80% ± 3.70%) (p < 0.001). Percentage change of myocardial blood flow in left anterior descending artery-dominated region, left ventricular ejection fraction, fractional shortening, and ST-segment resolution were higher, whereas infarcted area, troponin I, and creatine kinase MB isoenzyme were lower in ultrasound + microbubbles group than that in control group for both types of microthrombi (p < 0.001). Percentage change of myocardial blood flow, ejection fraction, fractional shortening, and ST-segment resolution were higher, whereas infarcted area, troponin I, and creatine kinase MB isoenzyme were lower in ultrasound + microbubbles and ultrasound + microbubbles + recombinant tissue-type plasminogen activator groups than that in recombinant tissue-type plasminogen activator group for platelet-rich microthrombi (p < 0.05). CONCLUSIONS: Ultrasound + microbubbles treatment could dissolve platelet- and erythrocyte-rich microthrombi, thereby improving outcomes of coronary no-reflow, making it a promising supplement to current reperfusion therapy for acute ST-segment elevation myocardial infarction.


Asunto(s)
Trombosis Coronaria/diagnóstico por imagen , Trombosis Coronaria/terapia , Microburbujas/uso terapéutico , Terapia Trombolítica/métodos , Animales , Modelos Animales de Enfermedad , Perros , Distribución Aleatoria , Resultado del Tratamiento , Ultrasonografía
9.
Front Cardiovasc Med ; 11: 1394929, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38932988

RESUMEN

Objective: Aging is the most significant contributor to the increasing prevalence of atrial fibrillation (AF). Dysbiosis of gut microbiota has been implicated in age-related diseases, but its role in AF development remains unclear. This study aimed to investigate the correlations between changes in the autonomic nervous system, short-chain fatty acids (SCFAs), and alterations in gut microbiota in aged rats with AF. Methods: Electrophysiological experiments were conducted to assess AF induction rates and heart rate variability in rats. 16S rRNA gene sequences extracted from fecal samples were used to assess the gut microbial composition. Gas and liquid chromatography-mass spectroscopy was used to identify SCFAs in fecal samples. Results: The study found that aged rats exhibited a higher incidence of AF and reduced heart rate variability compared to young rats. Omics research revealed disrupted gut microbiota in aged rats, specifically a decreased Firmicutes to Bacteroidetes ratio. Additionally, fecal SCFA levels were significantly lower in aged rats. Importantly, correlation analysis indicated a significant association between decreased SCFAs and declining heart rate variability in aged rats. Conclusions: These findings suggest that SCFAs, as metabolites of gut microbiota, may play a regulatory role in autonomic nervous function and potentially influence the onset and progression of AF in aged rats. These results provide novel insights into the involvement of SCFAs and autonomic nervous system function in the pathogenesis of AF. These results provide novel insights into the involvement of SCFAs and autonomic nervous system function in the pathogenesis of AF.

10.
Medicine (Baltimore) ; 103(4): e36799, 2024 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-38277535

RESUMEN

Pyroptosis plays a key role in the death of cells including cardiomyocytes, and it is associated with a variety of cardiovascular diseases. However, the role of pyroptosis-related genes (PRGs) in hypertrophic cardiomyopathy (HCM) is not well characterized. This study aimed to identify key biomarkers and explore the molecular mechanisms underlying the functions of the PRGs in HCM. The differentially expressed genes were identified by GEO2R, and the differentially expressed pyroptosis-related genes (DEPRGs) of HCM were identified by combining with PRGs. Enrichment analysis was performed using the "clusterProfiler" package of the R software. Protein-protein interactions (PPI) network analysis was performed using the STRING database, and hub genes were screened using cytoHubba. TF-miRNA coregulatory networks and protein-chemical interactions were analyzed using NetworkAnalyst. RT-PCR/WB was used for expression validation of HCM diagnostic markers. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western Blot (WB) were used to measure and compare the expression of the identified genes in the cardiac hypertrophy model and the control group. A total of 20 DEPRGs were identified, which primarily showed enrichment for the positive regulation of cytokine production, regulation of response to biotic stimulus, tumor necrosis factor production, and other biological processes. These processes primarily involved pathways related to Renin-angiotensin system, Adipocytokine signaling pathway and NF-kappa B signaling pathway. Then, a PPI network was constructed, and 8 hub genes were identified. After verification analysis, the finally identified HCM-related diagnostic markers were upregulated gene protein tyrosine phosphatase non-receptor type 11 (PTPN11), downregulated genes interleukin-1 receptor-associated kinase 3 (IRAK3), and annexin A2 (ANXA2). Further GSEA analysis revealed these 3 biomarkers primarily related to cardiac muscle contraction, hypertrophic cardiomyopathy, fatty acid degradation and ECM - receptor interaction. Moreover, we also elucidated the interaction network of these biomarkers with the miRNA network and known compounds, respectively. RT-PCR/WB results indicated that PTPN11 expression was significantly increased, and IRAK3 and ANXA2 expressions were significantly decreased in HCM. This study identified PTPN11, IRAK3, and ANXA2 as pyroptosis-associated biomarkers of HCM, with the potential to reveal the development and pathogenesis of HCM and could be potential therapeutic targets.


Asunto(s)
Cardiomiopatía Hipertrófica , MicroARNs , Humanos , Redes Reguladoras de Genes , Perfilación de la Expresión Génica/métodos , Piroptosis/genética , Biomarcadores , MicroARNs/genética , MicroARNs/metabolismo , Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/genética , Biología Computacional/métodos
11.
Eur J Pharmacol ; 965: 176307, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38160930

RESUMEN

OBJECTIVE: Inflammation and vascular smooth muscle cell (VSMC) phenotypic switching are implicated in the pathogenesis of abdominal aortic aneurysm (AAA). Trimethylamine N-oxide (TMAO) has emerged as a crucial risk factor in cardiovascular diseases, inducing vascular inflammation and calcification. We aimed to evaluate the effect of TMAO on the formation of AAA. APPROACH AND RESULTS: Here, we showed that TMAO was elevated in plasma from AAA patients compared with nonaneurysmal subjects by liquid chromatography‒mass spectrometry (LC‒MS) detection. Functional studies revealed that increased TMAO induced by feeding a choline-supplemented diet promoted Ang II-induced AAA formation. Immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), and Western blot analyses revealed that TMAO induced macrophage infiltration and inflammatory factor release. Conversely, inhibition of TMAO by supplementation with DMB suppressed AAA formation and the inflammatory response. Molecular studies revealed that TMAO regulated VSMC phenotypic switching. Flow cytometry analyses showed that TMAO induces macrophage M1-type polarization. Furthermore, pharmacological intervention experiments suggested that the nuclear factor-κB (NF-κB) signaling pathway was critical for TMAO to trigger AAA formation. CONCLUSIONS: TMAO promotes AAA formation by inducing vascular inflammation and VSMC phenotypic switching through activation of the NF-κB signaling pathway. Thus, TMAO is a prospective therapeutic AAA target.


Asunto(s)
Aneurisma de la Aorta Abdominal , Metilaminas , FN-kappa B , Humanos , Animales , FN-kappa B/metabolismo , Músculo Liso Vascular , Aneurisma de la Aorta Abdominal/patología , Inflamación/metabolismo , Miocitos del Músculo Liso , Angiotensina II/farmacología , Modelos Animales de Enfermedad
12.
Biomark Med ; 17(8): 417-426, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37489941

RESUMEN

Objective: The authors investigated the predictive value of MALAT1 for persistent atrial fibrillation (PAF) recurrence after radiofrequency ablation. Methods: Serum MALAT1 level was determined. The correlation between MALAT1 and high-sensitivity C-reactive protein/left atrial diameter (LAD) was analyzed. The predictive value of MALAT1 was evaluated. The postoperative recurrence rate in patients with high/low MALAT1 was compared. Independent risk factors for postoperative recurrence were analyzed. Results: MALAT1 was elevated in PAF patients and positively correlated with high-sensitivity C-reactive protein/LAD. MALAT1/high-sensitivity C-reactive protein/LAD were enhanced in patients with recurrent PAF. Patients with high MALAT1 had a higher recurrence rate. Upregulated MALAT1 was an independent risk factor for postoperative PAF recurrence. Conclusion: Serum MALAT1 level >2.03 predicts postoperative recurrence of PAF, and PAF patients with high MALAT1 have a higher risk of postoperative recurrence.


Atrial fibrillation (AF) is a common cardiac arrhythmia (abnormal heartbeat), which usually manifests as an irregular and rapid rhythm. In severe cases, AF can lead to cardiovascular diseases such as thromboembolism (narrowing and blockage of blood vessels) and heart failure (impaired pumping function of the heart). Cardiac radiofrequency ablation is a common method used to treat AF. However, patients with PAF still have a high rate of late recurrence after the operation, so there is an urgent need to identify suitable biochemical markers for predicting the postoperative recurrence of PAF. lncRNAs are a type of noncoding nucleic acid; they do not encode proteins, have various biological functions and are widely distributed in living organisms. The lncRNA MALAT1 has been considered a potential therapeutic target and biomarker in several cardiovascular diseases. This study demonstrated that the serum level of MALAT1 in PAF patients was significantly higher than that in normal subjects and MALAT1 level was elevated in patients with recurrent PAF compared with patients without recurrence. The authors also found that serum MALAT1 could predict whether PAF will recur after operation, with a high accuracy. In addition, PAF patients with high expression of serum MALAT1 had a higher risk of postoperative recurrence. In summary, serum level of lncRNA MALAT1 can help predict postoperative recurrence of PAF and a high level of MALAT1 is indicative of a higher risk of postoperative recurrence. Analysis of serum lncRNA MALAT1 level in PAF patients before surgery can predict postoperative recurrence, and intervention programs that lower the MALAT1 level can be implemented to reduce the risk of postoperative recurrence of PAF and increase the success rate of the operation. This study has important implications for reducing the recurrence rate after radiofrequency ablation in PAF patients.


Asunto(s)
Fibrilación Atrial , Ablación por Catéter , ARN Largo no Codificante , Ablación por Radiofrecuencia , Humanos , Fibrilación Atrial/genética , Fibrilación Atrial/cirugía , ARN Largo no Codificante/genética , Proteína C-Reactiva , Resultado del Tratamiento , Ablación por Catéter/efectos adversos , Factores de Riesgo , Recurrencia
14.
Front Cardiovasc Med ; 9: 1005306, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36187007

RESUMEN

In this study, Malus doumeri leaf extract (MDLE) was used to test its anti-oxidation capacity in vitro, it has been preliminarily analyzed for H2O2-induced oxidative damage in H9C2 cells and its main active components. The antioxidant capacity through DPPH (1, 1-Diphenyl-2-Picrylhydrazyl), ABTS+• [2,2,2'-azino-BIS-(3-ethylbenzo-thiazoline-6-sulfonic acid)] radical ion, •OH (hydroxyl radical), and • O 2 - (superoxide anion) were determined in vitro. The proliferation of H9C2 cells was examined by MTT [3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-Tetrazolium bromide]. MDA (malondialdehyde), SOD (superoxide dismutase), CAT (catalase), GSH (glutathione), and GSH-Px (glutathione peroxidase) were determined by colorimetry. Apoptosis induced by oxidative damage was detected by flow cytometry. The mRNA expression of antioxidant related genes of SOD, CAT, GSH, and GSH-Px were checked by qRT-PCR (quantitative real-time polymerase chain reaction). The MDLE main active components were analyzed by HPLC (high-performance liquid chromatography). MDLE had significant scavenging effects on DPPH, ABTS+•, •OH, and superoxide anion radicals in a concentration-dependent manner. H2O2 treatment could significantly lead to oxidative stress injury of H9C2 cells, and MDLE treatment significantly improved the degree of H9C2 cell damage, and showed a positive correlation with concentration. MDLE can also reduce apoptosis caused by oxidative damage. MDLE treatment could significantly reduce MDA content and increase CAT, SOD, GSH, and GSH-Px contents and expression. In addition, by HPLC analysis, the following six bioactive components were detected from MDLE: chlorogenic acid, isoquercitrin, quercetin, baicalin, and phloretin. Therefore, MDLE has a good protective effect on myocardial cells.

15.
Ann Transl Med ; 10(4): 168, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35280406

RESUMEN

Background: This study investigated the potential effects of 3-iodothyronamine (T1AM) on myocardial ischemia reperfusion injury (MIRI) and the underlying molecular mechanisms. Methods: A total of 16 adult male Sprague-Dawley rats were randomly divided into 4 groups and administered the following: control [60% dimethyl sulfoxide (DMSO) and 40% saline, pH 7.4], T1AM (25 mg/kg), T1AM (50 mg/kg), or T1AM (100 mg/kg). The rectal temperatures of the rats were measured at different time points. A further 30 adult male Sprague-Dawley rats were randomized and divided into the following 3 groups (n=10 in each group): sham operation, ischemia/reperfusion (I/R), and I/R + T1AM. In the I/R and I/R + T1AM groups, the left anterior descending (LAD) coronary artery of the rats were occluded for 0.5 hour to induce myocardial ischemia, followed by reperfusion for 3 hours in the I/R group. The electrocardiography (ECG), cardiac function, and 2,3,5-triphenyltetrazolium chloride (TTC) staining were examined in rats to evaluate the myocardial injury. The differences in the expression of apoptosis-related and Akt-FoxO1 signaling-related proteins were determined via Western blot. Results: This work verified that T1AM reduced the body temperature of rats in a dose-dependent manner. Additionally, T1AM improved cardiac function and decreased the infarction size caused by MIRI. T1AM reduced the expression of biochemical parameters and apoptosis of myocardial cells. In addition, after treatment with T1AM, the expression of Glut1, pFoxO1 and Akt were reduced, while the expression of FoxO1 and PPARα were increased significantly. Conclusions: Pretreatment of cardiomyocytes with T1AM inhibited apoptosis and protected against ischemia reperfusion injury via the Akt/FoxO1 signaling pathway.

16.
Life Sci ; 288: 119092, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33737086

RESUMEN

AIMS: Phenotypic switching of vascular smooth muscle cells (VSMCs) is essential for the formation of abdominal aortic aneurysms (AAAs). MicroRNA-23b (miR-23b) has recently been shown to play a vital role in maintaining the VSMC contractile phenotype; however, little is known about the role of miR-23b in the formation of AAAs. Here, we investigated whether miR-23b prevents AAA formation by inhibiting VSMC phenotypic switching. MATERIALS AND METHODS: We administered angiotensin II (Ang II, 1000 ng/kg/min) or vehicle to 10-12-week-old male apolipoprotein E knockout (ApoE-/-) or C57BL/6J mice via subcutaneous osmotic minipumps for 4 weeks. KEY FINDINGS: The expression of miR-23b was significantly reduced in the aorta during the early onset of AAA in angiotensin II-treated ApoE-/- mice and in human AAA samples. In vitro experiments showed that the suppression of SMC contractile marker gene expression induced by Ang II was accelerated by miR-23b inhibitors but inhibited by mimics. In vivo studies revealed that miR-23b deficiency in Ang II-treated C57BL/6J mice aggravated the formation of AAAs in these mice compared with control mice; the opposite results were observed in miR-23b-overexpressing mice. Mechanistically, miR-23b knockdown significantly increased the expression of the transcription factor forkhead box O4 (FoxO4) during VSMC phenotypic switching induced by Ang II. In addition, a luciferase reporter assay showed that FoxO4 is a target of miR-23b in VSMCs. SIGNIFICANCE: Our study revealed a pivotal role for miR-23b in protecting against aortic aneurysm formation by maintaining the VSMC contractile phenotype.


Asunto(s)
Angiotensina II/toxicidad , Aneurisma de la Aorta Abdominal/prevención & control , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/metabolismo , MicroARNs/genética , Miocitos del Músculo Liso/patología , Animales , Aneurisma de la Aorta Abdominal/etiología , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Proteínas de Ciclo Celular/genética , Factores de Transcripción Forkhead/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Miocitos del Músculo Liso/metabolismo , Fenotipo
17.
Int Urol Nephrol ; 53(11): 2321-2332, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33895976

RESUMEN

PURPOSE: Hyperuricemia (HUA) and hypertriglyceridemia (HTG) were very common in chronic kidney disease (CKD) and associated with accelerated progression of CKD. This was a retrospective, cross-sectional study which aimed to explore the relationship between serum uric acid levels or triglyceride levels and tubular atrophy/interstitial fibrosis (proven by renal biopsy). METHODS: The present study enrolled 229 CKD individuals who included 127 biopsy-proven primary IgA nephrology (IgAN) patients and 102 biopsy-proven primary membranous nephropathy (MN) patients. The baseline characteristics at the time of the kidney biopsy were collected. According to the serum uric acid (UA) or triglyceride (TG) whether it exceeds the normal reference range, patients were divided into non-HUA (n = 127), HUA (n = 102), non-HTG (n = 119), and HTG group (n = 110). Based on the extent of tubular atrophy/interstitial fibrosis, patients were divided into no/mild injury (T0, n = 127), moderate injury (T1, n = 102). Multivariable logistic regression for factors predicting moderate tubular atrophy/interstitial fibrosis was performed. RESULTS: There were 127 IgAN and 102 MN cases among 229 patients in the present study. The prevalence of HUA was 44.5% (n = 102), 40.9% (n = 52), and 49.0% (n = 50) in all patients, IgAN patients and MN patients, respectively (P = 1.49). The prevalence of HTG was 48.0% (n = 110), 29.9% (n = 38), and 70.6% (n = 72) (P < 0.001), respectively, as well. Multivariate logistic regression analysis showed that HUA and HTG were independent risk factors for moderate tubular atrophy/interstitial fibrosis (HUA OR = 2.335, 95% CI = 1.147-4.755, P = 0.019; HTG OR = 2.646, 95% CI = 1.289-5.432, P = 0.008). The area under curve (AUC) of model 1 (HUA + eGFR + HTG + age + serum globulin + 24 h urinary protein, AUC = 0.876) was larger than the other two models; however, there was no significant difference among these models (all P > 0.05). CONCLUSIONS: Hyperuricemia and hypertriglyceridemia, which were prevalent in CKD patients, were the independent risk factors for moderate tubular atrophy/interstitial fibrosis. HUA together with HTG could improve the value of diagnosis for moderate tubular atrophy/interstitial fibrosis to some extent.


Asunto(s)
Glomerulonefritis por IGA/complicaciones , Glomerulonefritis Membranosa/complicaciones , Hipertrigliceridemia/complicaciones , Hiperuricemia/complicaciones , Riñón/patología , Adulto , Atrofia/etiología , Estudios Transversales , Femenino , Fibrosis/etiología , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
18.
Mol Med Rep ; 23(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33179102

RESUMEN

Induction of cardiomyocyte (CM) proliferation is a promising approach for cardiac regeneration following myocardial injury. MicroRNAs (miRs) have been reported to regulate CM proliferation. In particular, miR­449a­5p has been identified to be associated with CM proliferation in previous high throughput functional screening data. However, whether miR­449a­5p regulates CM proliferation has not been thoroughly investigated. This study aimed to explore whether miR­449a­5p modulates CM proliferation and to identify the molecular mechanism via which miR­449a­5p regulates CM proliferation. The current study demonstrated that miR­449a­5p expression levels were significantly increased during heart development. Furthermore, the results suggested that miR­449a­5p mimic inhibited CM proliferation in vitro as determined via immunofluorescence for ki67 and histone H3 phosphorylated at serine 10 (pH3), as well as the numbers of CMs. However, miR­449a­5p knockdown promoted CM proliferation. CDK6 was identified as a direct target gene of miR­449a­5p, and CDK6 mRNA and protein expression was suppressed by miR­449a­5p. Moreover, CDK6 gain­of­function increased CM proliferation. Overexpression of CDK6 also blocked the inhibitory effect of miR­449a­5p on CM proliferation, indicating that CDK6 was a functional target of miR­449a­5p in CM proliferation. In conclusion, miR­449a­5p inhibited CM proliferation by targeting CDK6, which provides a potential molecular target for preventing myocardial injury.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina/genética , Corazón/crecimiento & desarrollo , MicroARNs/genética , Miocitos Cardíacos/citología , Regiones no Traducidas 3' , Animales , Proliferación Celular , Células Cultivadas , Quinasa 6 Dependiente de la Ciclina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Masculino , Ratones , Miocitos Cardíacos/química , Regulación hacia Arriba
19.
Mol Biol Rep ; 37(4): 1749-54, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19578979

RESUMEN

Proteinuria is a well-established exacerbating factor in chronic kidney disease. Although the mechanisms of albumin-induced tubulointerstitial damage have been extensively studied, the influence of mycophenolic acid (MPA) on tubular epithelial cells has not been sufficiently elucidated. MPA, the active metabolite of mycophenolate mofetil, is a potent, non-competitive, and reversible inhibitor of inosine-5'-monophosphate dehydrogenase, the rate-limiting enzyme for de novo purine synthesis. Monocyte chemoattractant protein 1 (MCP-1) is a 76-amino-acid chemokine thought to be the major chemotactic factor for monocytes. MCP-1 is found in macrophage-rich areas of atherosclerotic lesions. However, the mechanisms regulating MCP-1 expression by MPA in renal tubular epithelial cells were still unclear. In this study, the inhibitory effect of MPA on MCP-1 expression by albumin-induced renal tubular epithelial cells was investigated, and the roles of p38 mitogen-activated protein kinase (p38 MAPK) pathway were explored. MPA attenuated albumin-induced expression of MCP-1 mRNA and protein. The experiment suggested that MPA actively inhibited protein of MCP-1. The inhibitory effect of MPA on MCP-1 expression was mediated by the sequential attenuation of p38 MAPK expression. These inhibitory effects were partially inhibited by SB203580, a specific inhibitor of p38 MAPK. Taken together, these results suggest that the negative modulation of MCP-1 by MPA is partly dependent on p38 MAPK pathway.


Asunto(s)
Albúminas/farmacología , Quimiocina CCL2/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Túbulos Renales Proximales/citología , Ácido Micofenólico/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Línea Celular , Quimiocina CCL2/genética , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Imidazoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Piridinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
20.
Mol Ther Nucleic Acids ; 21: 394-411, 2020 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-32650237

RESUMEN

Identifying effective drugs to delay the progression of aortic aneurysms is a formidable challenge in vascular medicine. Methyltransferase-like 3 (METTL3) plays a key role in catalyzing the formation of N6-methyladenosine (m6A), but despite the functional importance of METTL3 and m6A in various fundamental biological processes, their roles in abdominal aortic aneurysm (AAA) are unknown. Here, we found that METTL3 knockdown in apolipoprotein E-deficient (ApoE-/-) mice treated with angiotensin II suppressed the formation of AAAs, while METTL3 overexpression exerted the opposite effects. Similar results were obtained in a calcium chloride (CaCl2)-induced mouse AAA model. Mechanistically, METTL3-dependent m6A methylation promoted primary microRNA-34a (miR-34a, pri-miR34a) maturation through DGCR8. Moreover, miR-34a overexpression significantly decreased SIRT1 expression and aggravated AAA formation, while miR-34a deficiency produced the opposite effects. In a rescue experiment, miR-34a knockdown or forced expression of SIRT1 partially attenuated the protective effects of METTL3 deficiency against AAA formation. Our studies reveal an important role for METTL3/m6A-mediated miR-34a maturation in AAA formation and provide a novel therapeutic target and diagnostic biomarker for AAA treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA