Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Lab Anal ; 36(1): e24083, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34850462

ABSTRACT

BACKGROUND: The current study aimed to evaluate the relationship between baseline serum lipoprotein (a) [Lp(a)] level and heart failure with reduced ejection fraction (HFrEF) development. METHODS: This was a retrospective study, and participants were enrolled from the outpatient clinic. All data were extracted from the electronic health record of the outpatient clinic system. The follow-up was performed through reviewing the clinical notes at the outpatient clinic system, and study outcome of the current study was the first diagnosis of HFrEF. Participants were divided into low Lp(a) (<30 mg/dl, n = 336) and high Lp(a) (≥30 mg/dl, n = 584) groups. RESULTS: Individuals in the high Lp(a) group were more likely to be men and have diabetes mellitus (DM) and dyslipidemia. Increased Lp(a) at baseline was positively associated with serum N-terminal pro-B natriuretic peptide level while negatively associated with left ventricular ejection fraction (LVEF) at follow-up. After adjusting for covariates, per 10 mg/dl increase in baseline Lp(a) remained significantly associated with HFrEF, with odds ratio of 1.17 (95% confidence interval of 1.05, 1.46). The magnitude of association between baseline Lp(a) level and HFrEF was greater in men and in individuals with DM or coronary heart disease (CHD), while it was weaker in individuals treated with beta-blocker at baseline. CONCLUSION: Increased Lp(a) at baseline was associated with HFrEF development. The adverse effects of Lp(a) were greater on men and individuals with DM or CHD, which were mitigated by beta-blocker therapy. These findings together underscore the possibility and usefulness of Lp(a) as a new risk factor to predict HFrEF.


Subject(s)
Heart Failure , Lipoprotein(a)/blood , Adult , Female , Heart Failure/blood , Heart Failure/epidemiology , Humans , Male , Middle Aged , Retrospective Studies
2.
Exp Cell Res ; 398(2): 112389, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33221316

ABSTRACT

Ischemia-reperfusion (I/R) injury is a multifactorial process triggered when an organ is subjected to transiently reduced blood supply. The result is a cascade of pathological complications and organ damage due to the production of reactive oxygen species following reperfusion. The present study aims to evaluate the role of activated calcium-sensing receptor (CaR)-cystathionine γ-lyase (CSE)/hydrogen sulfide (H2S) pathway in I/R injury. Firstly, an I/R rat model with CSE knockout was constructed. Transthoracic echocardiography, TTC and HE staining were performed to determine the cardiac function of rats following I/R Injury, followed by TUNEL staining observation on apoptosis. Besides, with the attempt to better elucidate how CaR-CSE/H2S affects I/R, in-vitro culture of human coronary artery endothelial cells (HCAECs) was conducted with gadolinium chloride (GdCl3, a CaR agonist), H2O2, siRNA against CSE (siCSE), or W7 (a CaM inhibitor). The interaction between CSE and CaM was subsequently detected. Plasma oxidative stress indexes, H2S and CSE, and apoptosis-related proteins were all analyzed following cell apoptosis. We found that H2S elevation led to the improvement whereas CSE knockdown decreased cardiac function in rats with I/R injury. Moreover, oxidative stress injury in I/R rats with CSE knockout was aggravated, while the increased expression of H2S and CSE in the aortic tissues resulted in alleviated the oxidative stress injury. Moreover, increased H2S and CSE levels were found to inhibit cell apoptotic ability in the aortic tissues after I/R injury, thus attenuating oxidative stress injury, accompanied by inhibited expression of apoptosis-related proteins. In HCAECs following oxidative stress treatment, siCSE and CaM inhibitor were observed to reverse the protection of CaR agonist. Coimmunoprecipitation assay revealed the interaction between CSE and CaM. Taken together, all above-mentioned data provides evidence that activation of the CaR-CSE/H2S pathway may confer a potent protective effect in cardiac I/R injury.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/metabolism , Protective Agents/metabolism , Receptors, Calcium-Sensing/metabolism , Reperfusion Injury/metabolism , Animals , Apoptosis , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Oxidative Stress , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology
3.
Postgrad Med ; 133(2): 195-201, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33131368

ABSTRACT

Objective: The current study was to evaluate the association of Lipoprotein (a) [Lp(a)] and in-hospital outcomes in patients with acute coronary syndrome (ACS) undergoing percutaneous coronary intervention (PCI).Methods: ACS patients undergoing PCI were retrospectively enrolled. Based on Lp(a) level, patients were divided into low (<30 mg/dL) and high (≥30 mg/dL) Lp(a) groups.Results: Compared to those with low Lp(a), patients with high Lp(a) had larger numbers of coronary arteries ≥70% stenosis and had longer coronary artery lesion (P < 0.05). After adjustment for covariates, high Lp(a) remained associated with higher odds of having coronary artery ≥70% stenosis, type C coronary lesion and pre-PCI TIMI flow grade 1/0. Patients with high Lp(a) had a higher unadjusted odds of acute stent thrombosis (odds ratio [OR] 1.10 and 95% confidence interval [CI] 1.01-2.27), congestive heart failure (OR 1.24 and 95% CI 1.15-2.38) and composite in-hospital outcomes (OR 1.28 and 95% CI 1.18-2.42). After adjustment for covariates, patients with high Lp(a) still had a higher odds of congestive heart failure (OR 1.08 and 95% CI 1.01-1.78) and composite in-hospital outcomes (OR 1.12 and 95% CI 1.04-1.81).Conclusion: In ACS patients undergoing PCI, compared to those with low Lp(a), patients with high Lp(a) had more severe coronary artery lesion, higher risk of congestive heart failure and composite in-hospital outcomes.


Subject(s)
Acute Coronary Syndrome , Coronary Artery Disease , Coronary Vessels , Heart Failure , Lipoprotein(a)/blood , Percutaneous Coronary Intervention , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/etiology , Acute Coronary Syndrome/physiopathology , Acute Coronary Syndrome/therapy , Aged , China/epidemiology , Coronary Artery Disease/blood , Coronary Artery Disease/complications , Coronary Artery Disease/diagnosis , Coronary Artery Disease/epidemiology , Coronary Vessels/diagnostic imaging , Coronary Vessels/pathology , Female , Heart Failure/diagnosis , Heart Failure/epidemiology , Hospitalization/statistics & numerical data , Humans , Male , Middle Aged , Outcome Assessment, Health Care , Percutaneous Coronary Intervention/adverse effects , Percutaneous Coronary Intervention/methods , Percutaneous Coronary Intervention/statistics & numerical data , Retrospective Studies , Risk Factors , Severity of Illness Index
4.
J Cell Physiol ; 234(4): 3634-3646, 2019 04.
Article in English | MEDLINE | ID: mdl-30191968

ABSTRACT

Myocardial ischemia and reperfusion injury (MIRI) includes major drawbacks, such as excessive formation of free radicals and also overload of calcium, which lead to cell death, tissue scarring, and remodeling. The current study aims to explore whether KRT1 silencing may ameliorate MIRI via the Notch signaling pathway in mouse models. Myocardial tissues were used for the determination of the positive rate of KRT1 protein expression, apoptosis of myocardial cells, creatine kinase (CK) and lactate dehydrogenase (LDH) expression, expression of related biomarkers as well as myocardial infarction area. The transfected myocardial cells were treated with KRT1-siRNA, Jagged1, and DAPT (inhibitor of Notch-1 signaling pathway). The expression of KRT1, NICD, Hes1, Bcl-2, and Bax protein was detected. The MTT assay was applied for cell proliferation and flow cytometry was used for cell apoptosis. Mice with MIRI had a higher positive rate of KRT1 protein expression, apoptosis of myocardial cells, CK and LDH expression, myocardial infarction area, increased expression of MDA, NO, SDH, IL-1, IL-6, TNF-α, CRP, KRT1, Bax protein, CK, and LDH, and decreased expression of SOD, NICD, Hes1, and Bcl-2. The downregulation of KRT1 led to decreased expression of KRT1 and Bax protein, increased expression of NICD, Hes1, and Bcl-2, decreased cell apoptosis, and improved cell proliferation. The inhibition of the Notch signaling pathway leads to reduced expression of Bax, increased expression of NICD, Hes1, and Bcl 2, and also decreased cell apoptosis and increased cell proliferation. Our data conclude that KRT1 silencing is able to make MIRI better by activating the Notch signaling pathway in mice.


Subject(s)
Gene Silencing , Keratin-1/genetics , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Receptors, Notch/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Inflammation Mediators/metabolism , Keratin-1/metabolism , Male , Mice, Inbred C57BL , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Oxidative Stress , Rats, Sprague-Dawley , Receptors, Notch/genetics , Signal Transduction
5.
Life Sci ; 208: 315-324, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-29857073

ABSTRACT

AIMS: Hydrogen sulfide (H2S) is a novel signaling molecule with potent cytoprotective actions. In this study, we hypothesize that exogenous H2S may protect cardiac cells against high glucose (HG)-induced myocardial injury and inflammation with the involvement of the CIRP-MAPK signaling pathway. MAIN METHODS: H9c2 cardiac cells cultured under HG conditions were transfected with siRNA and different inhibitor for detecting the effects of sodium hydrogen sulfide (NaHS) (a H2S donor) on cell biological processes. The cardiac cell viability and LDH activity were determined by CCK-8 and LDH kit. ELISA was employed to measure the levels of inflammatory factors, while 2',7'-dichlorofluorescein diacetate (DCFH-DA) to evaluate reactive oxygen species (ROS). Mitochondrial membrane potential (MMP) was identified by rhodamine 123 staining. TUNEL staining and Hoechst 33258 staining were employed to observe cardiac cell apoptosis. Besides, we determined the expression of CIRP-MAPK signaling pathway- and apoptosis-related factors by protein immunoblot analysis. KEY FINDINGS: HG culturing induced toxicity, LDH, higher level of inflammatory factors, ROS, MMP, and apoptosis in cardiac cells, attenuated the viability of cardiac cells, and activated the CIRP-MAPK signaling pathway. Notably, CIRP silencing aggravated the above condition. H2S or blockade of the MAPK signaling pathway reversed the above conditions induced by HG. SIGNIFICANCE: The present study provides evidence for the protective effect of exogenous H2S on HG-induced myocardial injury and inflammation in H9c2 cardiac cells and suggests that the activation of CIRP-MAPK signaling pathway might be one of the mechanisms underlying the protective effect of H2S.


Subject(s)
Cold Shock Proteins and Peptides/metabolism , Glucose/toxicity , Hydrogen Sulfide/pharmacology , Inflammation/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , RNA-Binding Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Cold Shock Proteins and Peptides/genetics , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Membrane Potential, Mitochondrial/drug effects , Myocardial Reperfusion Injury/chemically induced , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation/drug effects , Protective Agents/pharmacology , RNA-Binding Proteins/genetics , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/genetics
6.
Am J Physiol Cell Physiol ; 315(3): C380-C388, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29741915

ABSTRACT

Recent studies have uncovered the vital roles played by microRNAs in regulating cardiac injury. Among them, the cardiac enriched microRNA-1 (miR-1) has been extensively studied and proven to be detrimental to cardiac myocytes. Hence, the current study aimed to explore whether miR-1 affects myocardial ischemia-reperfusion injury (MIRI) in rats undergoing sevoflurane preconditioning and the underlying mechanism. After successful model establishment, rats with MIRI were transfected with mimics or inhibitors of miR-1, or siRNA against MAPK3, and then were injected with sevoflurane. A luciferase reporter gene assay was conducted to evaluate the targeting relationship between miR-1 and MAPK3. Reverse transcription quantitative polymerase chain reaction and Western blot analysis were employed to evaluate the expressions of miR-1, MAPK3, phosphatidylinositol 3-kinase (PI3K), and Akt. Additionally, the concentration of lactate dehydrogenase (LDH) was determined. Cell apoptosis and viability were assessed using TUNEL and cell counting kit-8 assays, and the ischemic area at risk and infarct size were detected using Evans blue and triphenyltetrazolium chloride staining. MAPK3 was found to be the target gene of miR-1. miR-1 expressed at a high level whereas MAPK3 expressed at a low level in MIRI rats. Overexpressing miR-1 or silencing MAPK3 blocked the PI3K/Akt pathway to increase cell apoptosis, ischemic area at risk, and infarct area but decreased cell viability and increased LDH concentration. In contrast, miR-1 downregulation abrogated the effects induced by miR-1 mimics or siRNA against MAPK3. These findings indicate that inhibition of miR-1 promotes MAPK3 to protect against MIRI in rats undergoing sevoflurane preconditioning through activation of the PI3K/Akt pathway.


Subject(s)
MicroRNAs/genetics , Mitogen-Activated Protein Kinases/genetics , Myocardial Reperfusion Injury/genetics , Phosphatidylinositol 3-Kinase/genetics , Proto-Oncogene Proteins c-akt/genetics , Sevoflurane/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Down-Regulation/drug effects , Down-Regulation/genetics , Ischemic Preconditioning, Myocardial/methods , L-Lactate Dehydrogenase/genetics , Male , Myocytes, Cardiac/drug effects , PC12 Cells , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...