Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Redox Biol ; 67: 102907, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37797372

RESUMEN

Cardiac fibrosis is characterized by the excessive deposition of extracellular matrix in the myocardium with cardiac fibroblast activation, leading to chronic cardiac remodeling and dysfunction. However, little is known about metabolic alterations in fibroblasts during cardiac fibrosis, and there is a lack of pharmaceutical treatments that target metabolic dysregulation. Here, we provided evidence that fatty acid ß-oxidation (FAO) dysregulation contributes to fibroblast activation and cardiac fibrosis. With transcriptome, metabolome, and functional assays, we demonstrated that FAO was downregulated during fibroblast activation and cardiac fibrosis, and that perturbation of FAO reversely affected the fibroblast-to-myofibroblast transition. The decrease in FAO may be attributed to reduced long-chain fatty acid (LCFA) uptake. Voltage-dependent anion channel 1 (VDAC1), the main gatekeeper of the outer mitochondrial membrane (OMM), serves as the transporter of LCFA into the mitochondria for further utilization and has been shown to be decreased in myofibroblasts. In vitro, the addition of exogenous VDAC1 was shown to ameliorate cardiac fibroblast activation initiated by transforming growth factor beta 1 (TGF-ß1) stimuli, and silencing of VDAC1 displayed the opposite effect. A mechanistic study revealed that VDAC1 exerts a protective effect by regulating LCFA uptake into the mitochondria, which is impaired by an inhibitor of carnitine palmitoyltransferase 1A. In vivo, AAV9-mediated overexpression of VDAC1 in myofibroblasts significantly alleviated transverse aortic constriction (TAC)-induced cardiac fibrosis and rescued cardiac function in mice. Finally, we treated mice with the VDAC1-derived R-Tf-D-LP4 peptide, and the results showed that R-Tf-D-LP4 prevented TAC-induced cardiac fibrosis and dysfunction in mice. In conclusion, this study provides evidence that VDAC1 maintains FAO metabolism in cardiac fibroblasts to repress fibroblast activation and cardiac fibrosis and suggests that the VDAC1 peptide is a promising drug for rescuing fibroblast metabolism and repressing cardiac fibrosis.


Asunto(s)
Fibroblastos , Canal Aniónico 1 Dependiente del Voltaje , Animales , Ratones , Ácidos Grasos/metabolismo , Fibroblastos/metabolismo , Fibrosis , Péptidos/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/genética , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
2.
Aging Dis ; 14(2): 261-282, 2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37008052

RESUMEN

Bile acids (BAs), key regulators in the metabolic network, are not only involved in lipid digestion and absorption but also serve as potential therapeutic targets for metabolic disorders. Studies have shown that cardiac dysfunction is associated with abnormal BA metabolic pathways. As ligands for several nuclear receptors and membrane receptors, BAs systematically regulate the homeostasis of metabolism and participate in cardiovascular diseases (CVDs), such as myocardial infarction, diabetic cardiomyopathy, atherosclerosis, arrhythmia, and heart failure. However, the molecular mechanism by which BAs trigger CVDs remains controversial. Therefore, the regulation of BA signal transduction by modulating the synthesis and composition of BAs is an interesting and novel direction for potential therapies for CVDs. Here, we mainly summarized the metabolism of BAs and their role in cardiomyocytes and noncardiomyocytes in CVDs. Moreover, we comprehensively discussed the clinical prospects of BAs in CVDs and analyzed the clinical diagnostic and application value of BAs. The latest development prospects of BAs in the field of new drug development are also prospected. We aimed to elucidate the underlying mechanism of BAs treatment in CVDs, and the relationship between BAs and CVDs may provide new avenues for the prevention and treatment of these diseases.

3.
JCI Insight ; 8(6)2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36787190

RESUMEN

Cardiac fibrosis is associated with an adverse prognosis in cardiovascular disease that results in a decreased cardiac compliance and, ultimately, heart failure. Recent studies have identified the role of long noncoding RNA (lncRNA) in cardiac fibrosis. However, the functions of many lncRNAs in cardiac fibrosis remain to be characterized. Through a whole-transcriptome sequencing and bioinformatics analysis on a mouse model of pressure overload-induced cardiac fibrosis, we screened a key lncRNA termed thrombospondin 1 antisense 1 (THBS1-AS1), which was positively associated with cardiac fibrosis. In vitro functional studies demonstrated that the silencing of THBS1-AS1 ameliorated TGF-ß1 effects on cardiac fibroblast (CF) activation, and the overexpression of THBS1-AS1 displayed the opposite effect. A mechanistic study revealed that THBS1-AS1 could sponge miR-221/222 to regulate the expression of TGFBR1. Moreover, under TGF-ß1 stimulation, the forced expression of miR-221/222 or the knockdown TGFBR1 significantly reversed the THBS1-AS1 overexpression induced by further CF activation. In vivo, specific knockdown of THBS1-AS1 in activated CFs significantly alleviated transverse aorta constriction-induced (TAC-induced) cardiac fibrosis in mice. Finally, we demonstrated that the human THBS1-AS1 can also affect the activation of CFs by regulating TGFBR1. In conclusion, this study reveals that lncRNA THBS1-AS1 is a potentially novel regulator of cardiac fibrosis and may serve as a target for the treatment of cardiac fibrosis.


Asunto(s)
Cardiomiopatías , MicroARNs , ARN Largo no Codificante , Humanos , Ratones , Animales , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , MicroARNs/genética , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Trombospondina 1/genética , Trombospondina 1/metabolismo , Fibrosis , Cardiomiopatías/metabolismo , Fibroblastos/metabolismo
4.
Pharmacol Res ; 188: 106677, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36702426

RESUMEN

Cardiac fibrosis is a pathological process underlying myocardial remodeling and is characterized by excessive deposition of the myocardial extracellular matrix. Long noncoding RNAs (lncRNAs) have emerged as critical regulators of various biological processes. In this study, we investigated the role of a novel lncRNA, Gm41724, in cardiac fibrosis induced by pressure overload. High-throughput whole transcriptome sequencing analysis was performed to detect differentially expressed lncRNAs in cardiac fibroblasts (CFs) with or without TGF-ß1 treatment. Differential expression analysis and gene set enrichment analysis identified Gm41724 as a potential molecule targeting fibrosis. Gm41724 positively regulated the activation of CFs induced by TGF-ß1 and pressure overload. Knocking down Gm41724 could inhibit the differentiation of CFs into myofibroblasts and alleviate cardiac fibrosis induced by pressure overload. Mechanistically, comprehensive identification of RNA-binding proteins by mass spectrometry (CHIRP-MS) and RNA immunoprecipitation (RIP) assay combined with other methods of molecular biological revealed the important role of Gm41724 binding to lamina-associated polypeptide 2α (lap2α) for the activation of CFs. Further mechanistic studies indicated that the regulator of G protein signaling 4 (Rgs4), as the downstream effector of Gm41724/lap2α, regulated CFs activation. Our results implicated the involvement of Gm41724 in cardiac fibrosis induced by pressure overload and it is expected to be a promising target for anti-fibrotic therapy.


Asunto(s)
Cardiomiopatías , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Cardiomiopatías/metabolismo , Miocardio/patología , Fibrosis , Fibroblastos/metabolismo
5.
Front Nutr ; 9: 948691, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35983482

RESUMEN

Background: Several studies have found a strong association between cardiovascular diseases and myeloperoxidase (MPO) as a marker of oxidative stress. Although the anti-inflammatory effects of vitamin D in adults have been validated, evidence about the relationship between MPO and 25(OH)D is lacking. This study aimed to investigate the relationship between MPO and 25(OH)D in the general Chinese population. Methods: From November 2018 to August 2019, a total of 6414 subjects were enrolled in a tertiary referral hospital in China, which included 3,122 women and 3,292 men. The dependent and independent variables were MPO and 25(OH)D, respectively. The confounders included age, sex, body mass index, waist-hip ratio, smoking status, alcohol drinking status, calcium, and parathyroid hormone concentration. Results: In the fully adjusted model, we found that MPO decreased by 0.12 (95% CI -0.16, -0.08), ng/mL for each unit (1 nmol/L) increase in 25(OH)D. When 25(OH) D was divided into quartiles, compared with Q1 (< 41.4 nmol/L), the adjusted beta coefficients (ß) of MPO in Q2-Q4 were -2.29 (95% CI, -4.31 to -0.27), -4.76 (95% CI, -6.83 to -2.69), and -6.07 (95% CI, -8.23 to -3.92), respectively (P for the trend < 0.0001). When 25(OH) D was divided according to clinical severity, compared with the severely deficient (< 30 nmol/L) s≥ 30, < 50 nmol/L) and sufficient groups (≥ 50 nmol/L) were -2.59 (95% CI, -5.87 to 0.69) and -5.87 (95% CI, -9.17 to -2.57), respectively (P for the trend < 0.0001). Conclusion: After adjusting for age, sex, BMI, waist-hip ratio, smoking status, alcohol status, calcium, and PTH, circulating 25(OH)D was negatively associated with MPO.

6.
Front Pharmacol ; 13: 940768, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36003513

RESUMEN

Cardiac hypertrophy initially serves as an adaptive response to physiological and pathological stimuli. Sustained hypertrophy progress to pathological cardiac hypertrophy, cardiac fibrosis and ultimately lead to heart failure, one of the leading medical causes of mortality worldwide. Intervention of pathological cardiac hypertrophy can effectively reduce the occurrence of heart failure. Abundant factors, such as adrenergic, angiotensin, and endothelin (ET-1) receptors, have been shown to participate in the regulation of pathological cardiac hypertrophy. Recently, an increasing number of studies have indicated that circRNA and circRNA-miRNA-mRNA network regulation is indispensable for the posttranscriptional regulation of mRNA in cardiac hypertrophy. In our study, the morphological, cardiac function and pathological changes during cardiac hypertrophy were investigated. RNA sequencing identified 93 circRNAs that were differentially expressed in the TAC_2w group, and 55 circRNAs in the TAC_4w group compared with the sham group. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses identified several significant pathways, including hypertrophic cardiomyopathy, extracellular matrix (ECM)-receptor interaction and focal adhesion. Coexpression analyses were performed for differentially expressed circRNAs and differentially expressed mRNAs. Based on gene set enrichment analysis (GSEA), 8 circRNAs (mmu-Nfkb1_0001, mmu-Smad4_0007, mmu-Hecw2_0009, mmu-Itgbl1_0002, mmu-Lrrc2_0005, mmu-Cpeb3_0007, mmu-Ryr2_0040, and mmu-Rtn4_0001) involved in cardiac hypertrophy and cardiac fibrosis were identified. We validated some key circRNAs by qPCR. The crucial coexpression of circRNA-mRNA and its interaction with miRNA showed the possible mechanism of circRNAs in the process of cardiac dysfunction. Our results may provide promising targets for the treatment of pathological cardiac hypertrophy and fibrosis.

7.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 38(3): 574-582, 2021 Jun 25.
Artículo en Chino | MEDLINE | ID: mdl-34180204

RESUMEN

Long non-coding RNA (lncRNA) Dnm3os plays a critical role in peritendinous fibrosis and pulmonary fibrosis, but its role in the process of cardiac fibrosis is still unclear. Therefore, we carried out study by using the myocardial fibrotic tissues obtained by thoracic aortic constriction (TAC) in an early study of our group, and the in vitro cardiac fibroblast activation model induced by transforming growth factor-ß1 (TGF-ß1). Quantitative real-time polymerase chain reaction (RT-qPCR), Western blot, and collagen gel contraction test were used to identify the changes of activation phenotype and the expression of Dnm3os in cardiac fibroblasts. Small interfering RNA was used to silence Dnm3os to explore its role in the activation of cardiac fibroblasts. The results showed that the expression of Dnm3os was increased significantly in myocardial fibrotic tissues and in the activated cardiac fibroblasts. And the activation of cardiac fibroblasts could be alleviated by Dnm3os silencing. Furthermore, the TGF-ß1/Smad2/3 pathway was activated during the process of cardiac fibroblasts activation, while was inhibited after silencing Dnm3os. The results suggest that Dnm3os silencing may affect the process of cardiac fibroblast activation by inhibiting TGF-ß1/Smad2/3 signal pathway. Therefore, interfering with the expression of lncRNA Dnm3os may be a potential target for the treatment of cardiac fibrosis.


Asunto(s)
ARN Largo no Codificante , Fibroblastos , Fibrosis , Humanos , Miocardio/patología , Transducción de Señal , Factor de Crecimiento Transformador beta1
8.
Front Pharmacol ; 12: 654425, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995071

RESUMEN

Purinergic P2X7 receptor, a nonselective cation channel, is highly expressed in immune cells as well as cardiac smooth muscle cells and endothelial cells. Its activation exhibits to mediate nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) inflammasome activation, resulting in the release of interleukin-1 beta (IL-1ß) and interleukin-18 (IL-18), and pyroptosis, thus triggering inflammatory response. These pathological mechanisms lead to the deterioration of various cardiovascular diseases, including atherosclerosis, arrhythmia, myocardial infarction, pulmonary vascular remodeling, and cardiac fibrosis. All these worsening cardiac phenotypes are proven to be attenuated after the P2X7 receptor inhibition in experimental studies. The present review aimed to summarize key aspects of P2X7 receptor-mediated inflammation and pyroptosis in cardiovascular diseases. The main focus is on the evidence addressing the involvement of the P2X7 receptor in the inflammatory responses to the occurrence and development of cardiovascular disease and therapeutic interventions.

9.
Front Pharmacol ; 12: 671809, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025431

RESUMEN

Mechanosensing and mechanotransduction are vital processes in mechanobiology and play critical roles in regulating cellular behavior and fate. There is increasing evidence that purinergic P2 receptors, members of the purinergic family, play a crucial role in cellular mechanotransduction. Thus, information on the specific mechanism of P2 receptor-mediated mechanotransduction would be valuable. In this review, we focus on purinergic P2 receptor signaling pathways and describe in detail the interaction of P2 receptors with other mechanosensitive molecules, including transient receptor potential channels, integrins, caveolae-associated proteins and hemichannels. In addition, we review the activation of purinergic P2 receptors and the role of various P2 receptors in the regulation of various pathophysiological processes induced by mechanical stimuli.

10.
Front Pharmacol ; 12: 627773, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679406

RESUMEN

Cardiac fibroblasts (CFs) activation is a hallmark feature of cardiac fibrosis caused by cardiac remodeling. The purinergic signaling molecules have been proven to participate in the activation of CFs. In this study, we explored the expression pattern of P2Y receptor family in the cardiac fibrosis mice model induced by the transverse aortic constriction (TAC) operation and in the activation of CFs triggered by transforming growth factor ß1 (TGF-ß1) stimulation. We then investigated the role of P2Y1receptor (P2Y1R) in activated CFs. The results showed that among P2Y family members, only P2Y1R was downregulated in the heart tissues of TAC mice. Consistent with our in vivo results, the level of P2Y1R was decreased in the activated CFs, when CFs were treated with TGF-ß1. Silencing P2Y1R expression with siP2Y1R accelerated the effects of TGF-ß1 on CFs activation. Moreover, the P2Y1R selective antagonist BPTU increased the levels of mRNA and protein of profibrogenic markers, such as connective tissue growth factor (CTGF), periostin (POSTN). periostin (POSTN), and α-smooth muscle actin(α-SMA). Further, MRS2365, the agonist of P2Y1R, ameliorated the activation of CFs and activated the p38 MAPK and ERK signaling pathways. In conclusion , our findings revealed that upregulating of P2Y1R may attenuate the abnormal activation of CFs via the p38 MAPK and ERK signaling pathway.

13.
Intern Emerg Med ; 16(3): 661-668, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32888111

RESUMEN

Arterial stiffness has been implicated in pathophysiology of heart failure (HF) since it is involved in the ventricular-vascular coupling. Recently, new indices obtained by a cuff oscillometric technique, the arterial velocity pulse index (AVI) for the stiffness of central arteries and the arterial pressure volume index (API) for the stiffness of peripheral arteries have been developed and validated. However, the AVI and API measurement has not been attempted in HF population. This study aimed to investigate the relationship between the AVI, API and clinical parameters and outcomes in HF patients. A prospective cohort of patients with acute decompensated HF were enrolled within 6 months, who were admitted to a tertiary referral hospital in China. Measurement of the AVI and API (AVE-1500, Shisei Datum, Tokyo, Japan) was performed on the day of admission and discharge. Patients were followed up to 6 months for the composite endpoint of all-cause death and rehospitalization for HF. A total of 127 patients were recruited for analysis (60 ± 15 years, 70% male). 80% of the patients were in New York Heart Association (NYHA) Class III or IV at admission with mean left ventricular ejection fraction (LVEF) of 34 ± 9%. During hospitalization, all patients received guideline-directed medical therapy if not contraindicated. The AVI (27.3 ± 5.0 vs. 28.6 ± 6.7, P = 0.002) and API (24.9 ± 4.9 vs. 26.0 ± 6.5, P = 0.05) were lower at discharge than at admission. By dividing the patients into mild to severe group based on systolic blood pressure (SBP) and LVEF or into tertiles according to the amino-terminal pro-brain natriuretic peptide (NT-proBNP), transmitral E velocity over mitral annular e' velocity (E/e' ratio), it was observed that the AVI increased with a higher level of NT-proBNP (P for trend < 0.001), a larger E/e' (P for trend < 0.001) and a lower LVEF (P for trend = 0.0183), while the API increased as the E/e' and systolic blood pressure became higher (both P for trend < 0.05). The improvement in AVI at discharge was correlated with LVEF (R = - 0.3024, P < 0.05) and NT-proBNP improvement (R = 0.3118, P < 0.05), while the change in API was positively correlated with SBP change (R = 0.3897, P < 0.001). In 6 months after discharge, there were 52 predefined events including 15 deaths and 44 rehospitalization for HF. Apart from the level of NT-proBNP, the AVI at discharge of ≥ 26 showed a trend of being associated with the composite outcome (HR 2.747, 95% CI 1.411-5.349, P < 0.001 for univariate analysis; HR 1.864, 95% CI 0.892-3.893, P = 0.09761 for multivariate analysis). New noninvasive arterial stiffness indices as the AVI and API reflected severity of illness and midterm prognosis in admitted HF patients. Further studies are warranted for understanding its mechanisms and developing clinical applications.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Análisis de la Onda del Pulso , Rigidez Vascular , Enfermedad Aguda , Biomarcadores/sangre , Arteria Braquial/fisiopatología , China , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad , Volumen Sistólico
14.
Clin Interv Aging ; 15: 1649-1664, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32982199

RESUMEN

PURPOSE: Atherosclerotic cardiovascular disease may share the risk factors for low bone mineral density (BMD), one of which is dyslipidemia. The association between serum cholesterol and BMD remains controversial. Thus, the correlation between serum lipids and BMD in women was explored in the current study. MATERIALS AND METHODS: This cross-sectional study included 1116 Chinese female participants. Serum samples were collected to evaluate total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and other laboratory markers. Dual-energy X-ray absorptiometry was used to assess lumbar spine, femoral neck, and total hip BMD. RESULTS: In the postmenopausal women, a non-linear relationship was detected between TC, LDL-C, HDL-C, and lumbar spine BMD. Using segmented linear regression, the inflection points were 5.86 mmol/L, 3.52 mmol/L, and 2.37 mmol/L, respectively. To the left of the inflection point, the higher the serum lipid level, the lower the value for lumbar spine BMD. To the right of the inflection point, the higher the serum level of TC and LDL-C, the higher the value for lumbar spine BMD. In the premenopausal women, the association between HDL-C and femoral neck BMD was non-linear. In addition, LDL-C had a positive association with BMD of the femoral neck and HDL-C had an inverse association with BMD of the femoral neck in postmenopausal women. CONCLUSION: In postmenopausal women, the relationship between TC, LDL-C, HDL-C, and lumbar spine BMD was non-linear. TC, LDL-C, and HDL-C were negatively associated with lumbar spine BMD when the values were less than 5.86 mmol/L, 3.52 mmol/L, and 2.37 mmol/L, respectively. The mechanisms of the association were unclear, and further research is warranted to clarify the relationship.


Asunto(s)
Densidad Ósea/fisiología , Enfermedades Óseas Metabólicas/sangre , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Absorciometría de Fotón , Anciano , Pueblo Asiatico , Enfermedades Óseas Metabólicas/diagnóstico por imagen , Estudios Transversales , Femenino , Cuello Femoral/diagnóstico por imagen , Humanos , Vértebras Lumbares/diagnóstico por imagen , Persona de Mediana Edad , Premenopausia/sangre
16.
Oxid Med Cell Longev ; 2020: 7956274, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32566102

RESUMEN

P2X7 purinergic receptor (P2X7R) has been implicated in several cardiovascular diseases. However, whether it regulates cardiac fibrosis remains elusive. Herein, its involvement in the development of cardiac fibrosis was examined using a transverse aortic constriction (TAC) mice model and cardiac fibroblasts (CFs) hyperstimulated by TGF-ß1 for 48 hours. Results showed that TAC and TGF-ß1 treatment increased the expression of P2X7R. Silencing of P2X7R expression with siP2X7R ameliorated TGF-ß1 effects on fibroblasts activation. Similarly, P2X7R inhibition by Brilliant Blue G (BBG) reduced mRNA and protein levels of profibrosis markers, while the P2X7R agonist BzATP accelerated the TGF-ß1-induced CFs activation. Moreover, it was found that TGF-ß1-induced CFs activation was mediated by the NLRP3/IL-1ß inflammasome pathway. BBG or siP2X7R treatment suppressed NLRP3/IL-1ß pathway signaling. In vivo, BBG significantly alleviated TAC-induced cardiac fibrosis, cardiac dysfunction, and NLRP3/IL-1ß activation. Collectively, our findings imply that suppressing P2X7R may limit cardiac fibrosis and abnormal activation of CFs.


Asunto(s)
Interleucina-1beta/metabolismo , Miocardio/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7/metabolismo , Animales , Citoprotección/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Masculino , Ratones Endogámicos C57BL , Modelos Cardiovasculares , Presión , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
17.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 37(3): 450-459, 2020 Jun 25.
Artículo en Chino | MEDLINE | ID: mdl-32597087

RESUMEN

Calnexin is a lectin-like molecular chaperone protein on the endoplasmic reticulum, mediating unfolded protein responses, the endoplasmic reticulum Ca 2+ homeostasis, and Ca 2+ signals conduction. In recent years, studies have found that calnexin plays a key role in the heart diseases. This study aims to explore the role of calnexin in the activation of cardiac fibroblasts. A transverse aortic constriction (TAC) mouse model was established to observe the activation of cardiac fibroblasts in vivo, and the in vitro cardiac fibroblasts activation model was established by transforming growth factor ß1 (TGFß1) stimulation. The adenovirus was respectively used to gene overexpression and silencing calnexin in cardiac fibroblasts to elucidate the relationship between calnexin and cardiac fibroblasts activation, as well as the possible underlying mechanism. We confirmed the establishment of TAC model by echocardiography, hematoxylin-eosin, Masson, and Sirius red staining, and detecting the expression of cardiac fibrosis markers in cardiac tissues. After TGFß1 stimulation, markers of the activation of cardiac fibroblast, and proliferation and migration of cardiac fibroblast were detected by quantitative PCR, Western blot, EdU assay, and wound healing assay respectively. The results showed that the calnexin expression was reduced in both the TAC mice model and the activated cardiac fibroblasts. The overexpression of calnexin relieved cardiac fibroblasts activation, in contrast, the silencing of calnexin promoted cardiac fibroblasts activation. Furthermore, we found that the endoplasmic reticulum stress was activated during cardiac fibroblasts activation, and endoplasmic reticulum stress was relieved after overexpression of calnexin. Conversely, after the silencing of calnexin, endoplasmic reticulum stress was further aggravated, accompanying with the activation of cardiac fibroblasts. Our data suggest that the overexpression of calnexin may prevent cardiac fibroblasts against activation by alleviating endoplasmic reticulum stress.


Asunto(s)
Calnexina , Fibroblastos , Corazón , Chaperonas Moleculares , Animales , Calnexina/fisiología , Estrés del Retículo Endoplásmico , Fibroblastos/fisiología , Corazón/fisiología , Ratones
18.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 51(3): 331-336, 2020 May.
Artículo en Chino | MEDLINE | ID: mdl-32543138

RESUMEN

OBJECTIVE: The purpose of this study is to investigate the injury of liver and kidney tissues in overload pressure induced cardiac hypertrophy/heart failure mice model and the changes of macrophage activation level. METHODS: 6-8 week-old C57BL/6 mice were subjected to transverse aortic constriction (TAC) surgery to establish the cardiac hypertrophy/heart failure mouse model induced by pressure overload, while the aortic was not ligated in the Sham group. At 4 weeks and 8 weeks after TAC, the mice of each group were subjected to echocardiography and blood collection. And mice were sacrificed to collect samples of the heart, liver, and kidney tissues. The contents of plasma alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TBil) and serum creatinine (Scr) in Sham group and two operation groups were determined. The histological changes of liver, heart and kidney tissues were observed by HE staining, and the expression of the marker of macrophage activation, F4/80 protein, was detected in the heart, liver and kidney tissue by immunohistochemical staining. RESULTS: Cardiac hypertrophy occurred at 4 weeks after TAC operation in C57BL/6 mice and developed into heart failure at 8 weeks after TAC. The echocardiography showed that, compared with the Sham group, the left ventricular end-diastolic posterior wall thickness (LVPWd) and the left ventricular internal diameter in diastole (LVIDd) were significantly increased, while the left ventricular ejection fraction (EF) and the left ventricular fractional shortening (FS) were significantly decreased ( P<0.05) in the 4-week-TAC group and 8-week-TAC group. The plasma content of ALT, AST, TBil and Scr in the 4-week-TAC group and 8-week-TAC group were significantly higher than those in the Sham group ( P<0.05). HE staining showed obvious liver pathological changes in TAC mice, such as vacuolation, mild hepatic sinusoid congestion and inflammatory infiltration in mice post 4 weeks after surgery, and such liver injury was worse in mice post 8 weeks after surgery. Besides, there was a slight damage in renal tissue shown by HE staining, such as slight glomerular injury and slight bleeding. F4/80 protein immunohistochemical staining results demonstrated that the activation of macrophages in the heart and liver in the 4-week-TAC group and 8-week-TAC group was significantly increased than that in the sham group ( P<0.05), but there was no significant difference in kidney tissues in groups. CONCLUSION: Macrophages are involved in the process of liver and kidney injury in cardiac hypertrophy/heart failure.


Asunto(s)
Cardiomegalia , Insuficiencia Cardíaca , Función Ventricular Izquierda , Animales , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/etiología , Riñón/lesiones , Hígado/lesiones , Macrófagos , Ratones , Ratones Endogámicos C57BL , Volumen Sistólico
19.
Oxid Med Cell Longev ; 2020: 9423593, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32308810

RESUMEN

Mitochondrial DNA (mtDNA) damage is associated with the development of cardiovascular diseases. Cardiac aging plays a central role in cardiovascular diseases. There is accumulating evidence linking cardiac aging to mtDNA damage, including mtDNA mutation and decreased mtDNA copy number. Current wisdom indicates that mtDNA is susceptible to damage by mitochondrial reactive oxygen species (mtROS). This review presents the cellular and molecular mechanisms of cardiac aging, including autophagy, chronic inflammation, mtROS, and mtDNA damage, and the effects of mitochondrial biogenesis and oxidative stress on mtDNA. The importance of nucleoid-associated proteins (Pol γ), nuclear respiratory factors (NRF1 and NRF2), the cGAS-STING pathway, and the mitochondrial biogenesis pathway concerning the development of mtDNA damage during cardiac aging is discussed. Thus, the repair of damaged mtDNA provides a potential clinical target for preventing cardiac aging.


Asunto(s)
Envejecimiento/metabolismo , Sistema Cardiovascular/metabolismo , ADN Mitocondrial/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos
20.
World J Stem Cells ; 11(12): 1104-1114, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31875871

RESUMEN

Stem cells have shown great potential in vascular repair. Numerous evidence indicates that mechanical forces such as shear stress and cyclic strain can regulate the adhesion, proliferation, migration, and differentiation of stem cells via serious signaling pathways. The enrichment and differentiation of stem cells play an important role in the angiogenesis and maintenance of vascular homeostasis. In normal tissues, blood flow directly affects the microenvironment of vascular endothelial cells (ECs); in pathological status, the abnormal interactions between blood flow and vessels contribute to the injury of vessels. Next, the altered mechanical forces are transduced into cells by mechanosensors to trigger the reformation of vessels. This process occurs when signaling pathways related to EC differentiation are initiated. Hence, a deep understanding of the responses of stem cells to mechanical stresses and the underlying mechanisms involved in this process is essential for clinical translation. In this the review, we provide an overview of the role of stem cells in vascular repair, outline the performance of stem cells under the mechanical stress stimulation, and describe the related signaling pathways.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA