Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Int Immunopharmacol ; 136: 112338, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38850787

ABSTRACT

Cardiac fibrosis is a typical feature of cardiac pathological remodeling, which is associated with adverse clinical outcomes and has no effective therapy. Nicotine is an important risk factor for cardiac fibrosis, yet its underlying molecular mechanism remains poorly understood. This study aimed to identify its potential molecular mechanism in nicotine-induced cardiac fibrosis. Our results showed nicotine exposure led to the proliferation and transformation of cardiac fibroblasts (CFs) into myofibroblasts (MFs) by impairing autophagy flux. Through the use of drug affinity responsive target stability (DARTS) assay, cellular thermal shift assay (CETSA), and surface plasmon resonance (SPR) technology, it was discovered that nicotine directly increased the stability and protein levels of lactate dehydrogenase A (LDHA) by binding to it. Nicotine treatment impaired autophagy flux by regulating the AMPK/mTOR signaling pathway, impeding the nuclear translocation of transcription factor EB (TFEB), and reducing the activity of cathepsin B (CTSB). In vivo, nicotine treatment exacerbated cardiac fibrosis induced in spontaneously hypertensive rats (SHR) and worsened cardiac function. Interestingly, the absence of LDHA reversed these effects both in vitro and in vivo. Our study identified LDHA as a novel nicotine-binding protein that plays a crucial role in mediating cardiac fibrosis by blocking autophagy flux. The findings suggest that LDHA could potentially serve as a promising target for the treatment of cardiac fibrosis.


Subject(s)
Autophagy , Fibrosis , Nicotine , Animals , Autophagy/drug effects , Rats , Male , Rats, Inbred SHR , Signal Transduction/drug effects , Myocardium/pathology , Myocardium/metabolism , Lactate Dehydrogenase 5/metabolism , Cells, Cultured , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , TOR Serine-Threonine Kinases/metabolism , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Rats, Sprague-Dawley
2.
Food Funct ; 15(12): 6359-6373, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38787699

ABSTRACT

The aim of this study was to evaluate the preventive role and underlying mechanisms of fucoxanthin (Fx) on lipopolysaccharide (LPS)-induced intestinal barrier injury in mice. Our results demonstrated that the oral administration of Fx (50 and 200 mg per kg body weight per day) for consecutive 7 days significantly alleviated the severity of LPS-induced intestinal barrier injury in mice, as evidenced by attenuating body weight loss, improving intestinal permeability, and ameliorating intestinal morphological damage such as reduction in the ratio of the villus length to the crypt depth (V/C), intestinal epithelium distortion, goblet cell depletion, and low mucin 2 (MUC2) expression. Fx also significantly mitigated LPS-induced excessive apoptosis of intestinal epithelial cells (IECs) and curbed the decrease of tight junction proteins including claudin-1, occludin, and zonula occludens-1 in the ileum and colon. Additionally, Fx effectively alleviated LPS-induced extensive infiltration of macrophages and neutrophils into the intestinal mucosa, the overproduction of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin 1beta (IL-1ß) and IL-6, and gasdermin D (GSDMD)-mediated pyroptosis of IECs. The underlying mechanisms might be associated with inhibiting the activation of nuclear factor-kappa B (NF-κB), mitogen-activated protein kinases (MAPKs) and nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome signaling pathways. Moreover, Fx also notably restrained intestinal reactive oxygen species (ROS), malondialdehyde and protein carbonylation levels in LPS-treated mice, and it might be mediated by activating the nuclear factor-erythroid 2 related factor 2 (Nrf2) signaling pathway. Overall, these findings indicated that Fx might be developed as a potential effective dietary supplement to prevent intestinal barrier injury.


Subject(s)
Intestinal Mucosa , Lipopolysaccharides , Xanthophylls , Animals , Mice , Xanthophylls/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipopolysaccharides/adverse effects , Male , Apoptosis/drug effects , NF-kappa B/metabolism , Permeability , Mice, Inbred C57BL , Tight Junction Proteins/metabolism , Cytokines/metabolism
3.
Cardiorenal Med ; 13(1): 344-353, 2023.
Article in English | MEDLINE | ID: mdl-37839394

ABSTRACT

INTRODUCTION: Due to the cardiotoxicity of cancer treatment and traditional risk factors for cardiovascular disease (CVD) such as obesity, diabetes, dyslipidemia, and hypertension, cancer patients are at higher risk of developing CVD. However, limited research exists on the correlation between chronic kidney disease (CKD) and CVD risk in cancer patients. METHODS: This cross-sectional study selected cancer patients aged ≥20 years from the National Health and Nutrition Examination Survey (NHANES) conducted from 2015 to 2020. Multivariable logistic regression was used to assess the association between CKD and CVD in cancer patients. Additionally, subgroup analyses were conducted to investigate the association among different groups of cancer patients. RESULTS: We included 1,700 adult cancer patients (52.53% were females). After multivariable adjustment for covariates including traditional CVD factors, CKD was significantly associated with CVD, with an odds ratio (95% confidence interval) and p value of 1.61 (1.18, 2.19) and 0.004. Subgroup analyses after multivariable adjustment showed a significant correlation between CKD and increased CVD risk in the following cancer patients: age ≥60 years, males, white ethnicity, and individuals with or without traditional CVD factors (obesity, diabetes, dyslipidemia, and hypertension). CONCLUSIONS: CKD remains a significant factor in the higher risk of CVD among adult cancer patients in the United States, even after adjustment for traditional CVD risk factors. Therefore, to reduce the risk of CVD in cancer patients, it is important to treat CKD as a non-traditional risk factor for CVD and actively manage it.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus , Dyslipidemias , Hypertension , Neoplasms , Renal Insufficiency, Chronic , Adult , Male , Female , Humans , United States/epidemiology , Cardiovascular Diseases/complications , Cardiovascular Diseases/epidemiology , Nutrition Surveys , Cross-Sectional Studies , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/epidemiology , Hypertension/complications , Hypertension/epidemiology , Obesity/complications , Obesity/epidemiology , Dyslipidemias/complications , Dyslipidemias/epidemiology , Neoplasms/complications , Neoplasms/epidemiology
4.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 44(4): 545-554, 2022 Aug.
Article in Chinese | MEDLINE | ID: mdl-36065685

ABSTRACT

Objective To screen out the key genes leading to diabetic cardiomyopathy by analyzing the mRNA array associated with diabetic cardiomyopathy in the GEO database. Methods The online tool GEO2R of GEO was used to mine the differentially expressed genes (DEG) in the datasets GSE4745 and GSE5606.R was used to draw the volcano map of the DEG,and the Venn diagram was established online to identify the common DEG shared by the two datasets.The clusterProfile package in R was used for gene ontology annotation and Kyoto encyclopedia of genes and genomes pathway enrichment of the DEG.GSEA was used for gene set enrichment analysis,and STRING for the construction of a protein-protein interaction network.The maximal clique centrality algorithm in the plug-in Cytohubba of Cytoscape was used to determine the top 10 key genes. The expression of key genes was studied in the primary cardiomyocytes of rats and compared between the normal control group and high glucose group. Results The expression of Pdk4,Ucp3,Hmgcs2,Asl6,and Slc2a4 was consistent with the array analysis results.The expression of Pdk4,Ucp3,and Hmgcs2 was up-regulated while that of Acsl6 and Slc2a4 was down-regulated in the cardiomyocytes stimulated by high glucose (25 mmol/L) for 72 h. Conclusion Pdk4,Ucp3,Hmgcs2,Asl6,and Slc2a4 may be associated with the occurrence and development of diabetic cardiomyopathy,and may serve as the potential biomarkers of diabetic cardiomyopathy.


Subject(s)
Diabetes Mellitus , Diabetic Cardiomyopathies , Animals , Computational Biology/methods , Diabetic Cardiomyopathies/genetics , Gene Expression Profiling , Glucose , Protein Interaction Maps/genetics , Rats
5.
Food Funct ; 13(7): 3853-3864, 2022 Apr 04.
Article in English | MEDLINE | ID: mdl-35274650

ABSTRACT

Acute liver injury is a life-threatening syndrome that often results from the actions of viruses, drugs and toxins. Herein, the protective effect and potential mechanism of krill oil (KO), a novel natural product rich in long-chain n-3 polyunsaturated fatty acids bound to phospholipids and astaxanthin, on lipopolysaccharide (LPS)-evoked acute liver injury in mice were investigated. Male C57BL/6J mice were administered intragastrically with 400 mg kg-1 KO or fish oil (FO) once per day for 28 consecutive days prior to LPS exposure (10 mg kg-1, intraperitoneally injected). The results revealed that KO pretreatment significantly ameliorated LPS-evoked hepatic dysfunction indicated by reduced serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities and attenuated hepatic histopathological damage. KO pretreatment also mitigated LPS-induced hepatic oxidative stress, as evidenced by decreased malondialdehyde (MDA) contents, elevated glutathione (GSH) levels, and elevated catalase (CAT) and superoxide dismutase (SOD) activities. Additionally, LPS-evoked overproduction of pro-inflammatory mediators in serum and the liver was inhibited by KO pretreatment. Furthermore, KO pretreatment suppressed LPS-induced activation of the hepatic toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB)/NOD-like receptor family pyrin domain containing 3 (NLRP3) signaling pathway. Interestingly, the hepatoprotective effect of KO was superior to that of FO. Collectively, the current findings suggest that KO protects against LPS-evoked acute liver injury via inhibition of oxidative stress and inflammation.


Subject(s)
Chemical and Drug Induced Liver Injury , Euphausiacea , Animals , Chemical and Drug Induced Liver Injury/metabolism , Euphausiacea/metabolism , Inflammation/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/toxicity , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Stress
6.
J Agric Food Chem ; 70(9): 2911-2922, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35174699

ABSTRACT

Intestinal barrier dysfunction has emerged as a potential contributor to the development of several severe diseases. Herein, the effect and underlying mechanism of DHA-enriched phospholipids (DHA-PL) and EPA-enriched phospholipids (EPA-PL) on protecting against lipopolysaccharide (LPS)-induced intestinal barrier injury were elucidated. C57BL/6J male mice were fed an AIN-93G diet containing 1% DHA-PL or EPA-PL for 4 weeks and then were intraperitoneally injected with LPS (10 mg/kg) to cause intestinal barrier injury. The results manifested that DHA-PL and EPA-PL pretreatment balanced apoptosis and autophagy in intestinal epithelial cells and maintained intestinal tight junction integrity. Our findings also demonstrated that cotreatment with EX-527, a sirtuin 1 specific inhibitor, hindered the role of DHA-PL and EPA-PL against LPS-evoked intestinal barrier injury through reversing the inhibitory action of them on NF-κB and MAPKs activation as well as their potentiating actions on Nrf2 nuclear translocation. Overall, DHA-PL and EPA-PL alleviated LPS-mediated intestinal barrier injury via inactivation of the NF-κB and MAPKs pathways as well as activating the Nrf2 antioxidant pathway via up-regulating sirtuin 1.


Subject(s)
Eicosapentaenoic Acid , Lipopolysaccharides , Animals , Docosahexaenoic Acids/pharmacology , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred C57BL , Phospholipids , Sirtuin 1/genetics , Sirtuin 1/metabolism
7.
Front Physiol ; 12: 650055, 2021.
Article in English | MEDLINE | ID: mdl-34177609

ABSTRACT

Nicotine contained in traditional cigarettes, hookahs, and e-cigarettes is an important risk factor for cardiovascular disease. Our previous study showed that macroautophagic flux impairment occurred under nicotine stimulation. However, whether nicotine influences mitochondrial dynamics in neonatal rat ventricular myocytes (NRVMs) is unclear. The purpose of this study was to explore the effects and potential mechanism of nicotine on mitophagy, mitochondrial dynamics, apoptosis, and the relationship between these processes in NRVMs. Our results showed that nicotine exposure increased mitochondria-derived superoxide production, decreased mitochondrial membrane potential, and impaired PINK1/Parkin-mediated mitophagic flux in NRVMs. Interestingly, nicotine significantly promoted dynamin-related protein 1 (Drp1)-mediated mitochondrial fission and suppressed mitofusin (MFN)-mediated fusion, which was also observed in the bafilomycin A1-treated group. These results suggest that mitophagic flux impairment may contribute to Drp-1-mediated mitochondrial fission. Finally, nicotine caused excessive mitochondrial fission and contributed to apoptosis, which could be alleviated by mdivi-1, an inhibitor of Drp1. In addition to CTSB, as we previously reported, the enzyme activity of cathepsin L (CTSL) was also decreased in lysosomes after stimulation with nicotine, which may be the main cause of the hindered mitophagic flux induced by nicotine in NRVMs. Pretreatment with Torin 1, which is an inhibitor of mTOR, activated CTSL and ameliorated nicotine-induced mTOR activation and mitophagy impairment, decreased mitochondria-derived superoxide production, and blunted mitochondrial fission and apoptosis. Pretreatment with the ROS scavenger N-acetyl-cysteine (NAC) or inhibitors of p38 and JNK, which could also alleviate mitophagy impairment, exhibited similar effects as Torin1 on mitochondria. Taken together, our study demonstrated that nicotine treatment may lead to an increase in Drp1-mediated mitochondrial fission by blocking mitophagic flux by weakening the enzyme activity of CTSL and activating the ROS/p38/JNK signaling pathway. Excessive mitochondrial fission induced by nicotine ultimately leads to apoptosis. Torin1 restored the decreased CTSL enzyme activity by removing excessive ROS and alleviated the effects of nicotine on mitophagic flux, mitochondrial dynamics, and apoptosis. These results may provide new evidence on the relationship between mitophagic flux and mitochondrial dynamics and new perspectives on nicotine's effects on mitochondrial dynamics in cardiomyocytes.

8.
Biochem Biophys Res Commun ; 553: 172-179, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33773140

ABSTRACT

BACKGROUND: Cardiac fibrosis will increase wall stiffness and diastolic dysfunction, which will eventually lead to heart failure. Asenapine maleate (AM) is widely used in the treatment of schizophrenia. In the current study, we explored the potential mechanism underlying the role of AM in angiotensin II (Ang II)-induced cardiac fibrosis. METHODS: Cardiac fibroblasts (CFs) were stimulated using Ang II with or without AM. Cell proliferation was measured using the cell counting kit-8 assay and the Cell-Light EdU Apollo567 In Vitro Kit. The expression levels of proliferating cell nuclear antigen (PCNA) and α-smooth muscle actin (α-SMA) were detected using immunofluorescence or western blotting. At the protein level, the expression levels of the components of the transforming growth factor beta 1 (TGFß1)/mitogen-activated protein kinase (MAPK) signaling pathway were also detected. RESULTS: After Ang II stimulation, TGFß1, TGFß1 receptor, α-SMA, fibronectin (Fn), collagen type I (Col1), and collagen type III (Col3) mRNA levels increased; the TGFß1/MAPK signaling pathway was activated in CFs. After AM pretreatment, cell proliferation was inhibited, the numbers of PCNA -positive cells and the levels of cardiac fibrosis markers decreased. The activity of the TGFß1/MAPK signaling pathway was also inhibited. Therefore, AM can inhibit cardiac fibrosis by blocking the Ang II-induced activation through TGFß1/MAPK signaling pathway. CONCLUSIONS: This is the first report to demonstrate that AM can inhibit Ang II-induced cardiac fibrosis by down-regulating the TGFß1/MAPK signaling pathway. In this process, AM inhibited the proliferation and activation of CFs and reduced the levels of cardiac fibrosis markers. Thus, AM represents a potential treatment strategy for cardiac fibrosis.


Subject(s)
Angiotensin II/pharmacology , Dibenzocycloheptenes/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , MAP Kinase Signaling System/drug effects , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Fibroblasts/cytology , Fibrosis/metabolism , Fibrosis/prevention & control , Myocardium/cytology , Myocardium/metabolism , Rats , Rats, Wistar , Schizophrenia/drug therapy
9.
Int J Biol Sci ; 16(11): 2001-2013, 2020.
Article in English | MEDLINE | ID: mdl-32398966

ABSTRACT

Nicotine is proved to be an important factor for cardiac hypertrophy. Autophagy is important cell recycling system involved in the regulation of cardiac hypertrophy. Cilostazol, which is often used in the management of peripheral vascular disease. However, the effects of cilostazol on nicotine induced autophagy and cardiac hypertrophy are unclear. Here, we aim to determine the role and molecular mechanism of cilostazol in alleviating nicotine-induced cardiomyocytes hypertrophy through modulating autophagy and the underlying mechanisms. Our results clarified that nicotine stimulation caused cardiomyocytes hypertrophy and autophagy flux impairment significantly in neonatal rat ventricular myocytes (NRVMs), which were evidenced by augments of LC3-II and p62 levels, and impaired autophagosomes clearance. Interestingly, cathepsin B (CTSB) activity decreased dramatically after stimulation with nicotine in NRVMs, which was crucial for substrate degradation in the late stage of autophagy process, and cilostazol could reverse this effect dramatically. Intracellular ROS levels were increased significantly after nicotine exposure. Meanwhile, p38MAPK and JNK were activated after nicotine treatment. By using ROS scavenger N-acetyl-cysteine (NAC) could reverse the effects of nicotine by down-regulation the phosphorylation of p38MAPK and JNK pathways, and pretreatment of specific inhibitors of p38MAPK and JNK could restore the autophagy impairment and cardiomyocytes hypertrophy induced by nicotine. Moreover, CTSB activity of lysosome regained after the treatment with cilostazol. Cilostazol also inhibited the ROS accumulation and the activation of p38MAPK and JNK, which providing novel connection between lysosome CTSB and ROS/p38MAPK/JNK related oxidative stress pathway. This is the first demonstration that cilostazol could alleviate nicotine induced cardiomyocytes hypertrophy through restoration of autophagy flux by activation of CTSB and inhibiting ROS/p38/JNK pathway, exhibiting a feedback loop on regulation of autophagy and cardiomyocytes hypertrophy.


Subject(s)
Cathepsin B/metabolism , Cilostazol/pharmacology , Myocytes, Cardiac/drug effects , Nicotine/toxicity , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Bronchodilator Agents/pharmacology , Cathepsin B/genetics , Gene Expression Regulation/drug effects , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Nicotinic Agonists/toxicity , Proto-Oncogene Proteins/genetics , Rats , Receptor Protein-Tyrosine Kinases/genetics , p38 Mitogen-Activated Protein Kinases/genetics
10.
Int Heart J ; 60(4): 899-909, 2019 Jul 27.
Article in English | MEDLINE | ID: mdl-31308326

ABSTRACT

To systematically review and conduct a meta-analysis of the ivabradine-induced improvement in cardiopulmonary function, exercise capacity, and primary composite endpoints in patients with chronic heart failure (CHF).This study was a systematic review and meta-analysis.Databases, including PubMed, Embase, Cochrane Central Register of Controlled Trials (CENTRAL), and Clinical Trials and European Union Clinical Trials, were searched for randomized placebo-controlled trials. The efficacy and safety of ivabradine treatment in patients with CHF were assessed and compared to those of the standard anti-heart failure treatment. Review Manager 5.3 software was used to analyze the relative risk (RR) for dichotomous data and the mean difference (MD) for continuous data.In total, 22 studies with 24,562 patients were included. Cardiopulmonary function analysis showed that treatment with added ivabradine reduced the heart rate (MD = -17.30, 95% confidence interval (CI): 19.52--15.08, P < 0.00001), significantly increased the left ventricular ejection fraction (LVEF) (MD = 3.90, 95% CI: 0.40-7.40, P < 0.0001), and led to a better New York Heart Association (NYHA) classification. Ivabradine significantly reduced the minute ventilation/carbon dioxide production (VE/VCO2) (MD = -2.68, 95% CI: -4.81--0.55, P = 0.01) and improved the peak VO2 (MD = 2.80, 95% CI: 1.05-4.55, P = 0.002) and the exercise capacity, including the exercise duration with a submaximal load (MD = 7.82, 95% CI: -2.57--18.21, P < 0.00001) and the 6-minute walk distance. The RR of cardiovascular death or worsening heart failure was significantly decreased (RR = 0.93, 95% CI: 0.87--0.98, P = 0.01) in the patients treated with ivabradine. Additionally, the RRs of heart failure and hospitalization also decreased (RR = 0.91, 95% CI: 0.85--0.97, P = 0.006; RR = 0.86, 95% CI: 0.79--0.93, P = 0.0002). Safety analysis showed no significant difference in the RR of severe adverse events between the ivabradine group and the standard anti-heart failure treatment group (P = 0.40). However, ivabradine significantly increased the RR of visual symptoms in CHF patients (RR = 3.82, 95% CI: 1.80--8.13, P = 0.0005).Existing evidence showed that adding ivabradine treatment significantly improved the cardiopulmonary function and increased the exercise capacity of patients with CHF. Adding ivabradine to the standard anti-heart failure treatment reduced the mortality and hospitalization risk and improved the quality of life. Finally, ivabradine significantly increased the RR of visual symptoms in CHF patients.This is the first systematic review and meta-analysis to focus on the efficacy of ivabradine, which improved the cardiac function and increased the exercise capacity in patients with chronic heart failure (CHF). Therefore, this study will help evaluate the quality of life after adding ivabradine to the treatment of patients with CHF, even though there are differences in the standard for resting heart rate, left ventricular ejection fraction (LVEF), and New York Heart Association (NYHA) class in the included studies. This hybrid effect might be smaller when analyzed separately but might have a higher heterogeneity when analyzed in multiple studies.


Subject(s)
Exercise Tolerance/drug effects , Heart Failure/drug therapy , Ivabradine/therapeutic use , Stroke Volume/physiology , Ventricular Function, Left/physiology , Cardiovascular Agents/therapeutic use , Heart Failure/physiopathology , Humans , Stroke Volume/drug effects , Ventricular Function, Left/drug effects
11.
Saudi J Biol Sci ; 25(6): 1016-1021, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30174496

ABSTRACT

Atorvastatin (ATV) may support mesenchymal stem cells (MSC) survival in post-infarct myocardium (MI) as inflammatory reactions, oxidative stress and hypoxia condition get started in such tissues after damage. However, limited aqueous insolubility and rapid first-pass metabolism reduce the systemic availability of ATV. The aim of the present investigation was to develop ATV loaded nanoparticles (ATVNPs) which might ensure the maximum availability of ATV in systemic circulation for longer duration and to strengthen the support to MSC survival. ATVNPs were synthesized using double emulsion solvent evaporation method and characterized as spherical shape, positive charged, nanoparticles of uniform size distribution and higher entrapment efficiency. ATVNPs were non-cytotoxic and showed sustained release (up to 28 days). Assessment of cardiac function (in terms of echocardiographic and left heart catheterization parameters) and cytokines estimation revealed efficient improvement in post-infarct myocardium condition of rat. In conclusion, ATVNP was developed successfully that may ensure safe, cost effective, and efficacious treatment of post-infarct myo-cardium when compared with that of MSC alone and MSC supplemented with ATV solution.

12.
Clin Cardiol ; 41(11): 1446-1454, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30225843

ABSTRACT

BACKGROUND: Although switching between ticagrelor and clopidogrel is common in clinical practice, the efficacy and safety of this de-escalation remain controversial. HYPOTHESIS: We assessed the occurrences, reasons, and outcomes of switching from ticagrelor to clopidogrel in patients with ST-segment elevation myocardial infarction (STEMI) undergoing successful primary percutaneous coronary intervention (PCI). METHODS: A total of 653 patients with STEMI were randomly assigned to receive loading dose of ticagrelor or clopidogrel before PCI and then received maintenance dose, respectively, for 12 months follow-up. The primary outcome was major adverse cardiac events (MACE), including cardiovascular death, nonfatal myocardial infarction, and stroke. The secondary outcome included unexpected rehospitalization for angina, coronary revascularization, and stent thrombosis. The safety outcome was bleeding described by the Bleeding Academic Research Consortium (BARC) criteria. RESULTS: A total of 602 participants completed the study. The rate of switching from ticagrelor to clopidogrel was 48.6% and the main reason was financial burden. The rate of secondary ischemic events in the de-escalation group was higher than that in the ticagrelor group (15.1% vs 5.6%, P = 0.008), but lower than that in the clopidogrel group (15.1% vs 24.6%, P = 0.03), while there were no significant differences in MACE among the three groups (P = 0.16). De-escalation, ticagrelor, and clopidogrel did not cause significant differences in the rates of major bleeding among the three groups (BARC ≥ 2, P = 0.34). CONCLUSION: Switching from ticagrelor to clopidogrel is very common in patients with STEMI in China. De-escalation might be safe but associated with high risk of ischemic events as compared to ticagrelor.


Subject(s)
Clopidogrel/administration & dosage , Drug Substitution , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/administration & dosage , ST Elevation Myocardial Infarction/surgery , Ticagrelor/administration & dosage , Aged , China , Clopidogrel/adverse effects , Clopidogrel/economics , Coronary Thrombosis/etiology , Coronary Thrombosis/therapy , Drug Costs , Drug Substitution/adverse effects , Drug Substitution/mortality , Female , Hemorrhage/chemically induced , Humans , Male , Middle Aged , Patient Readmission , Percutaneous Coronary Intervention/adverse effects , Percutaneous Coronary Intervention/mortality , Platelet Aggregation Inhibitors/adverse effects , Prospective Studies , Recurrence , Risk Factors , ST Elevation Myocardial Infarction/blood , ST Elevation Myocardial Infarction/economics , ST Elevation Myocardial Infarction/mortality , Stroke/etiology , Stroke/mortality , Ticagrelor/adverse effects , Ticagrelor/economics , Time Factors , Treatment Outcome
13.
Medicine (Baltimore) ; 97(16): e0254, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29668577

ABSTRACT

BACKGROUND: The non-steroidal mineralocorticoid receptor antagonist finerenone (BAY 94-8862) has been used to treat chronic heart failure (CHF) with reduced ejection fraction (HFrEF). However, conflicting results were reported for its efficacy and safety. The study aimed to compare the efficacy and safety of finerenone versus spironolactone or eplerenone in patients with chronic heart failure. METHODS: Electronic databases including MEDLINE, EMBASE, and CENTRAL were searched from inception to December 2017 for randomized controlled trials assessing finerenone treatment in patients with chronic heart failure. Data concerning the study's design, patients' characteristics, and outcomes were extracted. Risk ratio (RR) and mean differences (MD) were calculated using either fixed or random effects models. RESULTS: Three trials with 1520 CHF patients were included in the systematic review. In terms of anti-ventricular remodeling, we calculated the effective number of cases with a 30% reduction in NT-proBNP. Finerenone was equivalent to the existing steroidal mineralocorticoid antagonist (P < .05). However, the efficacy of finerenone appeared to be dose-dependent. At a dose of 10 mg/d finerenone was found to be marginally better than that of steroidal mineralocorticoid receptor antagonists (MRAs) (RR = 1.18, 95% confidence interval [CI] 0.88, 1.57, P > .05). The incidence of treatment-related adverse events (TEAEs) of finerenone at 10 mg/d was significantly lower than 25 to 50 mg/d of steroidal MRAs (RR = 0.81, 95% CI = 0.66-0.99, P = .04). Moreover, the serum potassium levels in the finerenone 10 mg/d group were lower than those in the 25 to 50 mg/d steroidal MRAs group (MD = -0.14, 95% CI -0.30-0.02, P = .09), whereas the estimated glomerular filtration rate (eGFR) was higher in finerenone versus steroidal MRAs treated patients (MD = 2.07, 95% CI -0.04-4.17, P = .05). CONCLUSIONS: Finerenone reduced NT-proBNP level, urinary albumin/creatinine ratio (UACR), and other biochemical indicators, in a dose-dependent manner. In terms of anti-ventricular remodeling in patient with chronic heart failure, finerenone at 10 mg/d is as effective as 20 to 50 mg/d of steroidal MRAs. However, finerenone is much safer to patients with chronic kidney disease.


Subject(s)
Heart Failure/drug therapy , Naphthyridines/pharmacology , Dose-Response Relationship, Drug , Drug Monitoring/methods , Heart Failure/diagnosis , Humans , Mineralocorticoid Receptor Antagonists/pharmacology , Treatment Outcome
14.
Acta Pharmacol Sin ; 38(4): 488-497, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28216620

ABSTRACT

Aspirin (ASA) is a cardioprotective drug with anti-cardiac fibrosis action in vivo. This study was aimed to clarify the anti-cardiac fibrosis action of ASA and the underlying mechanisms. Two heart injury models (injection of isoproterenol and ligation of the left anterior descending branch) were used in mice to induce cardiac fibrosis. The animals were treated with ASA (10 mg·kg-1·d-1, ig) for 21 and 14 d, respectively. ASA administration significantly improved cardiac function, and ameliorated heart damage and fibrosis in the mice. The mechanisms underlying ASA's anti-fibrotic effect were further analyzed in neonatal cardiac fibroblasts (CFs) exposed to hypoxia in vitro. ASA (0.5-5 mmol/L) dose-dependently inhibited the proliferation and Akt phosphorylation in the CFs. In addition, ASA significantly inhibited CF apoptosis, and decreased the levels of apoptosis markers (cleaved caspase 3 and Parp1), which might serve as a side effect of anti-fibrotic effect of ASA. Furthermore, ASA dose-dependently inhibited the autophagy in the CFs, as evidenced by the reduced levels of autophagy marker LC3-II. The autophagy inhibitor Pepstatin A (PepA) promoted the inhibitory effect of ASA on CF proliferation, whereas the autophagy inducer rapamycin rescued ASA-caused inhibition of CF proliferation, suggesting an autophagy-dependent anti-proliferative effect of ASA. Both p38 inhibitor SB203580 and ROS scavenger N-acetyl-cysteine (NAC) significantly decreased Akt phosphorylation in CFs in the basal and hypoxic situations, but they both significantly increased LC3-II levels in the CFs. Our results suggest that an autophagy- and p38/ROS-dependent pathway mediates the anti-cardiac fibrosis effect of ASA in CFs. As PepA and SB203580 did not affect ASA-caused inhibition of CF apoptosis, the drug combination will enhance ASA's therapeutic effects.


Subject(s)
Aspirin/therapeutic use , Autophagy/drug effects , Cardiomyopathies/drug therapy , Cardiotonic Agents/therapeutic use , Animals , Apoptosis/drug effects , Aspirin/pharmacology , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cardiotonic Agents/pharmacology , Cell Hypoxia , Fibroblasts/drug effects , Fibroblasts/pathology , Fibrosis/drug therapy , Fibrosis/pathology , Imidazoles/pharmacology , Mice, Inbred BALB C , Mice, Inbred C57BL , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
15.
Oncotarget ; 8(18): 30455-30463, 2017 May 02.
Article in English | MEDLINE | ID: mdl-27458166

ABSTRACT

Proprotein convertase-subtilisin/kexin type 9 (PCSK9) monoclonal antibody is a new therapy to reduce low-density lipoprotein cholesterol (LDL-C) level in patients with familial hypercholesterolemia (FH). This pooled analysis aimed to estimate the efficacy and safety of PCSK9 antibody therapy in FH. Reports of randomized controlled trials (RCTs) comparing PCSK9 antibody to placebo were retrieved by a search of MEDLINE via PubMed, EMBASE, the Cochrane Library databases, ClinicalTrials.gov and Clinical Trial Results (up to November 30, 2015) with no language restriction. Data were abstracted by a standardized protocol. We found eight RCTs (1,879 patients with FH) for the pooled analysis. As compared with placebo, PCSK9 antibody therapy remarkably reduced LDL-C level (mean reduction: -48.54 %, 95 % CI: -53.19 to -43.88), total cholesterol (mean reduction: -31.08%, 95 % CI: -35.20 to -26.95), lipoprotein (a) (mean reduction: -20.44%, 95 % CI: -25.21 to -15.66), and apolipoprotein B (mean reduction: -36.32%, 95 % CI: -40.75 to -31.90) and elevated the level of high-density lipoprotein cholesterol (mean change: 6.29 %, 95 % CI: 5.12 to 7.46) and apolipoprotein A1(mean change: 4.86%, 95 % CI: 3.77 to 5.95). Therapy with and without PCSK9 antibodies did not differ in rate of adverse events (pooled rate: 50.86 % vs. 48.63%; RR: 1.03; 95 % CI: 0.92 to 1.15; P = 0.64; heterogeneity P = 0.13; I2= 40%) or serious adverse events (pooled rate: 7.14% vs. 6.74%; RR: 1.05; 95 % CI: 0.70 to 1.58; P = 0.80; heterogeneity P = 0.69; I2= 0%). PCSK9 antibody may be an effective and safe treatment for FH.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hyperlipoproteinemia Type II/drug therapy , PCSK9 Inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Biomarkers , Clinical Trials as Topic , Humans , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/metabolism , Lipids/blood , Publication Bias , Treatment Outcome
16.
Biomed Pharmacother ; 83: 930-935, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27522255

ABSTRACT

Patchouli alcohol (PA) is a tricyclic sesquiterpene extracted from a traditional Chinese herb pogostemonis herba. Literatures have proven that PA could inhibit inflammatory responses in various inflammatory disease models. However, whether PA could protect against atherosclerosis, a chronic vascular inflammation, is unknown. In this study, we sought to explore this issue in atherosclerosis-prone apolipoprotein E knockout mice fed an atherogenic diet, with or without daily PA intragastrical administration (40mg/kg). Our results showed that PA administration did not change plasma lipids metabolism, however, it significantly attenuated atherosclerotic plaque burdens in both the aorta and the aortic root. The lesional macrophage content, shown as Mac2 positive areas, was reduced, while the lesional smooth muscle cell and collagen content, shown as α-SMA positive areas and by Sirius red staining, respectively, was not affected in PA-treated mice, compared with non-treated controls. Aortic mRNA expression of macrophage inflammatory cytokines, including MCP-1, iNOS, IL-1ß, IL-6, CXCL9 and CXCL11, was also reduced in PA-treated mice. Therefore, we demonstrated that PA could attenuate atherosclerosis, possibly by inhibiting macrophage infiltration and its inflammatory responses.


Subject(s)
Atherosclerosis/drug therapy , Inflammation/pathology , Macrophages/pathology , Sesquiterpenes/therapeutic use , Animals , Aorta/drug effects , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Gene Expression Regulation/drug effects , Inflammation/genetics , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Lipids/blood , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Knockout , Plaque, Atherosclerotic/pathology , Sesquiterpenes/pharmacology
17.
PLoS One ; 9(3): e90096, 2014.
Article in English | MEDLINE | ID: mdl-24594984

ABSTRACT

OBJECTIVES: MicroRNA-7 (miR-7) is highly connected to cancerous cell proliferation and metastasis. It is also involved in myocardial ischemia-reperfusion (I/R) injury and is upregulated in cardiomyocyte under simulated I/R (SI/R). We aimed to investigate the role of miR-7 during myocardial I/R injury in vitro and in vivo and a possible gene target. METHODS AND RESULTS: Real-time PCR revealed that miR-7a/b expression was upregulated in H9c2 cells after SI/R. Flow cytometry showed SI/R-induced cell apoptosis was decreased with miR-7a/b mimic transfection but increased with miR-7a/b inhibitor in H9c2 cells. In a rat cardiac I/R injury model, infarct size determination and TUNEL assay revealed that miR-7a/b mimic decreased but miR-7a/b inhibitor increased cardiac infarct size and cardiomyocyte apoptosis as compared with controls. We previously identified an important gene connected with cell apoptosis--poly(ADP-ribose) polymerase (PARP)--as a candidate target for miR-7a/b and verified the target by luciferase reporter activity assay and western blot analysis. CONCLUSIONS: miR-7a/b is sensitive to I/R injury and protects myocardial cells against I/R-induced apoptosis by negatively regulating PARP expression in vivo and in vitro. miR-7a/b may provide a new therapeutic approach for treatment of myocardial I/R injury. Poly(ADP-ribose) polymerase.


Subject(s)
MicroRNAs/physiology , Myocytes, Cardiac/pathology , Poly(ADP-ribose) Polymerases/metabolism , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Base Sequence , Cell Line , Humans , In Situ Nick-End Labeling , Myocytes, Cardiac/enzymology , Rats , Reverse Transcriptase Polymerase Chain Reaction
18.
PLoS One ; 8(12): e71466, 2013.
Article in English | MEDLINE | ID: mdl-24312440

ABSTRACT

BACKGROUND: Atherosclerosis is a chronic inflammatory disease mediated by immune cells. Th22 cells are CD4(+) T cells that secret IL-22 but not IL-17 or IFN-γ and are implicated in the pathogenesis of inflammatory disease. The roles of Th22 cells in the pathophysiologic procedures of acute coronary syndrome (ACS) remain unclear. The purpose of this study is to investigate the profile of Th22, Th17 and Th17/Th1 cells in ACS patients, including unstable angina (UA) and acute myocardial infarction (AMI) patients. DESIGN AND METHODS: In this study, 26 AMI patients, 16 UA patients, 16 stable angina (SA) patients and 16 healthy controls were included. The frequencies of Th22, Th17 and Th17/Th1 cells in AMI, UA, SA patients and healthy controls were examined by flow cytometry. Plasma levels of IL-22, IL-17 and IFN-γ were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS: Th22, Th17 and Th17/Th1 cells were significantly increased in AMI and UA patients compared with SA patients and healthy controls. Moreover, plasma IL-22 level was significantly elevated in AMI and UA patients. In addition, Th22 cells correlated positively with IL-22 as well as Th17 cells in AMI and UA patients. CONCLUSION: Our findings showed increased frequencies of both Th22 and Th17 cells in ACS patients, which suggest that Th22 and Th17 cells may play a potential role in plaque destabilization and the development of ACS.


Subject(s)
Acute Coronary Syndrome/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/pathology , Aged , CD4 Lymphocyte Count , Female , Humans , Interferon-gamma/blood , Interferon-gamma/immunology , Interleukins/blood , Interleukins/immunology , Male , Middle Aged , Th1 Cells/metabolism , Th1 Cells/pathology , Th17 Cells/metabolism , Th17 Cells/pathology , Interleukin-22
19.
Curr Med Res Opin ; 27(9): 1771-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21806315

ABSTRACT

BACKGROUND: Some clinical and experimental studies have shown the use of statins could protect against AF, but there are not adequate data at present. OBJECTIVES: We performed a meta-analysis of randomized trials with statins on the endpoint of incidence of AF to estimate the impact of statin use on AF development. METHODS: We searched PUBMED, EMBASE and the Cochrane controlled Trials Register (Cochrane Library Issue 4, 2010) up to November 2010 to identify studies covering the use of statins on atrial fibrillation. RESULTS: In published data from nine short term trials (1044 patients, 421 AF), the effect of statins was significantly associated with a decreased risk of recurrence of AF (OR 0.43, 95% CI 0.25 to 0.73, P = 0.002). The result of OR was higher when studies with Jadad score ≤3 were excluded (OR 0.32, 95% CI 0.18 to 0.54, P ≤ 0.0001). Among four long term trials (12,442 patients, 618 AF), the effect of statins was associated with a decreased risk of recurrence of AF (OR 0.81, 95% CI 0.68 to 0.97, P = 0.02). In three long term trials of more intensive versus standard statin (9130 patients, 188 AF), there was no evidence of a reduction in the risk of AF (OR 1.05, 95% CI 0.79 to 1.40, P= 0.74). CONCLUSION: Our meta-analysis suggests that the use of statins may be associated with preventing AF in short term trials and long term trials, but in the long term trials of more intensive versus standard statin, there was no evidence of a reduction in the risk of AF. However, we still need large-scale randomized double blind statin trials with AF occurrence as the main endpoint in order to finally confirm the benefits of statin in AF patients.


Subject(s)
Atrial Fibrillation/prevention & control , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Randomized Controlled Trials as Topic/statistics & numerical data , Algorithms , Atrial Fibrillation/epidemiology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Incidence , Publications/statistics & numerical data , Recurrence , Treatment Outcome
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 23(2): 195-7, 2006 Apr.
Article in Chinese | MEDLINE | ID: mdl-16604496

ABSTRACT

OBJECTIVE: To study the linkage between K469E polymorphism of intercellular adhesion molecule 1(ICAM1) gene with ICAM1 plasma level and coronary heart disease (CHD) in Han population of China. METHODS: One hundred and sixty-four controls without CHD and 160 patients with CHD were enrolled in our study. By nested PCR with allele-specific oligonucleotide primers, all patients and controls were genotyped for the ICAM1 polymorphism. And the ICAM1 plasma level was measured by ELISA. RESULTS: In the patients with CHD, both K allele frequency and the plasma level of ICAM1 were higher than those in control (P<0.05). The individual with K allele had higher plasma level of ICAM1 than that without K allele (344.34+/-128.59 microg/L vs 303.54+/-108.74 microg/L, P=0.008). K allele enhanced the risk of CHD (P<0.01, OR=2.158, 95%CI: 1.250-3.727). There was the K allele cooperation with smoking in influencing the risk of CHD. CONCLUSION: There is the polymorphism of ICAM1 K469E gene in Han population of China, and the K allele may be a genetic factor influencing the risk of CHD.


Subject(s)
Coronary Disease/blood , Intercellular Adhesion Molecule-1/genetics , Polymorphism, Genetic , China/ethnology , Coronary Disease/genetics , Gene Frequency , Humans , Intercellular Adhesion Molecule-1/blood , Potassium/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...