Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1355149, 2024.
Article in English | MEDLINE | ID: mdl-38745945

ABSTRACT

Objective: The baseline urinary albumin/creatinine ratio (uACR) has been proven to be significantly associated with the risk of major adverse cardiac events (MACE). However, data on the association between the longitudinal trajectory patterns of uACR, changes in glycated hemoglobin A1c (HbA1c), and the subsequent risk of MACE in patients with diabetes are sparse. Methods: This is a retrospective cohort study including 601 patients with type 2 diabetes mellitus (T2DM; uACR < 300 mg/g) admitted to The First Hospital of Shanxi Medical University and The Second Hospital of Shanxi Medical University from January 2015 to December 2018. The uACR index was calculated as urinary albumin (in milligrams)/creatinine (in grams), and latent mixed modeling was used to identify the longitudinal trajectory of uACR during the exposure period (2016-2020). The deadline for follow-up was December 31, 2021. The primary outcome was the MACE [a composite outcome of cardiogenic death, hospitalization related to heart failure (HHF), non-fatal acute myocardial infarction, non-fatal stroke, and acute renal injury/dialysis indications]. The Kaplan-Meier survival analysis curve was used to compare the risk of MACE among four groups, while univariate and multivariate Cox proportional hazards models were employed to calculate the hazard ratio (HR) and 95% confidence interval (CI) for MACE risk among different uACR or HbA1c trajectory groups. The predictive performance of the model, both before and after the inclusion of changes in the uACR and HbA1c, was evaluated using the area under the receiver operating characteristic (ROC) curve (AUC). Results: Four distinct uACR trajectories were identified, namely, the low-stable group (uACR = 5.2-38.3 mg/g, n = 112), the moderate-stable group (uACR = 40.4-78.6 mg/g, n = 229), the high-stable group (uACR = 86.1-153.7 mg/g, n = 178), and the elevated-increasing group (uACR = 54.8-289.4 mg/g, n = 82). In addition, five distinct HbA1c trajectories were also identified: the low-stable group (HbA1c = 5.5%-6.8%, n = 113), the moderate-stable group (HbA1c = 6.0%-7.9%, n = 169), the moderate-decreasing group (HbA1c = 7.4%-6.1%, n = 67), the high-stable group (HbA1c = 7.7%-8.9%, n = 158), and the elevated-increasing group (HbA1c = 8.4%-10.3%, n = 94). Compared with the low-stable uACR group, patients in the high-stable and elevated-increasing uACR groups were more likely to be older, current smokers, and have a longer DM course, higher levels of 2-h plasma glucose (PG), HbA1c, N-terminal pro-B-type natriuretic peptide (NT-proBNP), uACR, and left ventricular mass index (LVMI), while featuring a higher prevalence of hypertension and a lower proportion of ß-receptor blocker treatment (p < 0.05). During a median follow-up of 45 months (range, 24-57 months), 118 cases (19.6%) of MACE were identified, including 10 cases (1.7%) of cardiogenic death, 31 cases (5.2%) of HHF, 35 cases (5.8%) of non-fatal acute myocardial infarction (AMI), 18 cases (3.0%) of non-fatal stroke, and 24 cases (4.0%) of acute renal failure/dialysis. The Kaplan-Meier survival curve showed that, compared with that in the low-stable uACR group, the incidence of MACE in the high-stable (HR = 1.337, 95% CI = 1.083-1.652, p = 0.007) and elevated-increasing (HR = 1.648, 95% CI = 1.139-2.387, p = 0.009) uACR groups significantly increased. Similar results were observed for HHF, non-fatal AMI, and acute renal injury/dialysis indications (p < 0.05). The multivariate Cox proportional hazards models indicated that, after adjusting for potential confounders, the HRs for the risk of MACE were 1.145 (p = 0.132), 1.337 (p = 0.007), and 1.648 (p = 0.009) in the moderate-stable, high-stable, and elevated-increasing uACR groups, respectively. In addition, the HRs for the risk of MACE were 1.203 (p = 0.028), 0.872 (p = 0.024), 1.562 (p = 0.033), and 2.218 (p = 0.002) in the moderate-stable, moderate-decreasing, high-stable, and elevated-increasing groups, respectively. The ROC curve showed that, after adding uACR, HbA1c, or both, the AUCs were 0.773, 0.792, and 0.826, which all signified statistically significant improvements (p = 0.021, 0.035, and 0.019, respectively). Conclusion: A long-term elevated uACR is associated with a significantly increased risk of MACE in patients with diabetes. This study implies that regular monitoring of uACR could be helpful in identifying diabetic patients with a higher risk of MACE.


Subject(s)
Albuminuria , Creatinine , Diabetes Mellitus, Type 2 , Glycated Hemoglobin , Humans , Male , Female , Retrospective Studies , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/urine , Diabetes Mellitus, Type 2/blood , Middle Aged , Albuminuria/urine , Creatinine/urine , Creatinine/blood , Aged , Glycated Hemoglobin/analysis , Longitudinal Studies , Risk Factors , Prognosis , Biomarkers/urine , Biomarkers/blood , Cohort Studies , Follow-Up Studies
2.
Korean J Physiol Pharmacol ; 28(3): 285-294, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38682176

ABSTRACT

Myocardial infarction is one of the leading causes of mortality globally. Currently, the pleiotropic inflammatory cytokine interleukin-6 (IL-6) is considered to be intimately related to the severity of myocardial injury during myocardial infarction. Interventions targeting IL-6 are a promising therapeutic option for myocardial infarction, but the underlying molecular mechanisms are not well understood. Here, we report the novel role of IL-6 in regulating adverse cardiac remodeling mediated by fibroblasts in a mouse model of myocardial infarction. It was found that the elevated expression of IL-6 in myocardium and cardiac fibroblasts was observed after myocardial infarction. Further, fibroblast-specific knockdown of Il6 significantly attenuated cardiac fibrosis and adverse cardiac remodeling and preserved cardiac function induced by myocardial infarction. Mechanistically, the role of Il6 contributing to cardiac fibrosis depends on signal transduction and activation of transcription (STAT)3 signaling activation. Additionally, Stat3 binds to the Il11 promoter region and contributes to the increased expression of Il11, which exacerbates cardiac fibrosis. In conclusion, these results suggest a novel role for IL-6 derived from fibroblasts in mediating Stat3 activation and substantially augmented Il11 expression in promoting cardiac fibrosis, highlighting its potential as a therapeutic target for cardiac fibrosis.

3.
Front Physiol ; 14: 1161612, 2023.
Article in English | MEDLINE | ID: mdl-37228823

ABSTRACT

Objectives: Circulating exosomal microRNAs (miRNAs) have been identified as promising biomarkers for diagnosis of cardiovascular diseases. Nevertheless, the diagnostic potential of miRNAs in circulating exosomes for stable coronary artery disease (SCAD) remains unclear. We aim here to analyze the exosomal differentially expressed miRNAs (DEmiRNAs) in plasma of SCAD patients and investigate their diagnostic potential as SCAD biomarkers. Methods: Plasma was collected from SCAD patients and healthy controls, and exosomes were isolated by ultracentrifugation. Exosomal DEmiRNAs were analyzed by small RNA sequencing and were further validated by quantitative real-time PCR (qRT-PCR) in a larger set of plasma samples. Relationships between plasma exosomal let-7c-5p, miR-335-3p, miR-652-3p, genders and Gensini Scores in patients with SCAD were analyzed using correlation analyses. Moreover, we conducted receiver operating characteristic (ROC) curves for these DEmiRNAs and analyzed their possible functions and signaling pathways. Results: Vesicles isolated from plasma displayed all characteristics of exosomes. In the small RNA sequencing study, a total of 12 DEmiRNAs were identified, among which seven were verified to be statistically significant by qRT-PCR. The areas under the ROC curves of exosomal let-7c-5p, miR-335-3p, and miR-652-3p were 0.8472, 0.8029, and 0.8009, respectively. Exosomal miR-335-3p levels were positively correlated with Gensini scores of patients with SCAD. Bioinformatics analysis revealed that these DEmiRNAs may be involved in the pathogenesis of SCAD. Conclusion: Our findings indicated that plasma exosomal let-7c-5p, miR-335-3p, and miR-652-3p can be used as promising biomarkers for diagnosis of SCAD. In addition, plasma exosomal miR-335-3p levels coordinated with severity of SCAD.

4.
Front Oncol ; 13: 1030599, 2023.
Article in English | MEDLINE | ID: mdl-36816975

ABSTRACT

Objective: To explore the corrective effect of 6 degree of freedom couch on rotation errors in intensity modulated radiotherapy (IMRT) for postoperative rectal cancer patients, further to probe into the clinical application value of 6 degree of freedom couch in radiotherapy. Methods: From January 1, 2020 to December 1, 2020, 30 patients with rectal cancer receiving postoperative intensity modulated radiotherapy in The First Hospital of Hebei Medical University were included in this retrospective study. The setup error values in all direction of patients before and after 6 degree of freedom correction were collected during each radiotherapy session. Results: In this study, a total of 382 data before and after the correction of 6 degree of freedom couch were collected. It was found that the setup errors in the Y direction gradually increased, was maximal in the third week, and then became smaller, and the setup errors in the other directions increased with the extension of radiotherapy time and reached the maximum at the 5th week. In the translation direction, the setup errors value in Z direction occurred more frequently than that in X and Y directions between the range of 0.21-0.80 cm. In the rotation direction, the setup errors value in rotation X direction occurred more frequently than that in rotation Y and Z directions between the range of 0.21°-2.99°. In addition, after the correction of the 6 degree of freedom couch in real time, the setup errors in patients were significantly reduced in all directions (P < 0.05). Conclusion: In summary, it was recommended to clinically use 6 degree of freedom couch combined with IMRT for real-time correction of placement errors in patients with rectal cancer undergoing radiotherapy. At the same time, it was necessary to observe the tumor size and body weight changes of patients on the 5th week. If necessary, radiotherapy positioning and planning should be performed in time.

5.
Curr Med Chem ; 29(15): 2652-2672, 2022.
Article in English | MEDLINE | ID: mdl-34525914

ABSTRACT

Hyperinsulinism-hyperammonemia syndrome (HHS) is a rare disease characterized by recurrent hypoglycemia and persistent elevation of plasma ammonia, and it can lead to severe epilepsy and permanent brain damage. It has been demonstrated that functional mutations of glutamate dehydrogenase (GDH), an enzyme in the mitochondrial matrix, are responsible for the HHS. Thus, GDH has become a promising target for the small molecule therapeutic intervention of HHS. Several medicinal chemistry studies are currently aimed at GDH, however, to date, none of the compounds reported has been entered clinical trials. This perspective summarizes the progress in the discovery and development of GDH inhibitors, including the pathogenesis of HHS, potential binding sites, screening methods, and research models. Future therapeutic perspectives are offered to provide a reference for discovering potent GDH modulators and encourage additional research that will provide more comprehensive guidance for drug development.


Subject(s)
Hyperammonemia , Hyperinsulinism , Hypoglycemia , Glutamate Dehydrogenase/chemistry , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase/metabolism , Humans , Hyperammonemia/drug therapy , Hyperammonemia/genetics , Hyperinsulinism/drug therapy , Hyperinsulinism/genetics , Mutation
6.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 38(5): 504-509, 2022 Sep.
Article in Chinese | MEDLINE | ID: mdl-37088760

ABSTRACT

OBJECTIVE: To investigate the effects of p53 upregulated modulator of apoptosis (PUMA) on the apoptosis of H9C2 cardiomyocytes induced by high glucose and its mechanisms. METHODS: H9C2 cardiomyocytes were treated with 5.5mmol/L (control group) or 35 mmol/L glucose (HG group) for 6 h, 12 h, 24 h or 48 h respectively to induce apoptosis, each group sets 5 multiple wells. Apoptosis was tested by TUNEL assay. PUMA mRNA was measured by RT-PCR and protein expression was measured by Western blot assay. The mitochondrial membrane potential was detected by JC-1 method. The expressions of cleaved caspase-3 and cytochrome C (Cyt C) protein in mitochondria and cytoplasm were determined by Western blot assay. H9C2 cardiomyocytes were randomly divided into four groups, control group (5.5 mmol/L), HG (35 mmol/L) group, HG+si-scramble group(si-scramble treatment for 24 h, then 35 mmol/L high glucose treatment for 24 h) and HG-si-PUMA group (si-PUMA treatment for 24 h, then 35mmol/L high glucose treatment for 24 h). Si-PUMA was transfected into cardiomyocytes and the effects of PUMA on high glucose-induced apoptosis were studied. RESULTS: Compared with the control group, high glucose increased cardiomyocyte apoptosis and enhanced PUMA mRNA and protein expressions significantly (P<0.05 or P<0.01). Cell injury and increased PUMA expression were time-dependent and there was no significant difference between the high glucose 24 h group and the high glucose 48h group. The following experiment used high glucose 24 h as the stimulation time. The cardiomyocytes transfected with si-PUMA to inhibit PUMA expression had decreased apoptotic rate and cleaved caspase-3, increased mitochondria membrane potential and decreased Cyt C release (P<0.05 or P<0.01). There were no significant differences between the HG+si-scramble group and the high glucose group (P>0.05). CONCLUSION: PUMA mediates high glucose-induced cardiomyocyte apoptosis suggesting PUMA may be an important target gene of diabetic cardiomyopathy.


Subject(s)
Apoptosis , Myocytes, Cardiac , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Glucose/adverse effects , Glucose/metabolism , Myocytes, Cardiac/metabolism , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Rats
7.
Bioengineered ; 12(2): 11716-11725, 2021 12.
Article in English | MEDLINE | ID: mdl-34895040

ABSTRACT

This study was to explore the effects of oxidized-low density lipoprotein (ox-LDL) on the proliferation and apoptosis of macrophages, and the role of miRNA-9 in the targeted regulation of platelet-derived growth factor receptor-ß (PDGFR-ß) expression. Macrophage RAW264.7 cells were cultured and foamed with 100 mg/L ox-LDL to detect the cell proliferation and apoptosis and target protein expression levels. Subsequently, the miRNA-9 mimics and inhibitors were transfected to detect the expression level of PDGFR-ß. The dual-luciferase reporter gene was predicted and applied to detect the target-binding effect of miRNA-9 and PDGFR-ß in the cells. The results showed that ox-LDL could induce the foaming of macrophages RAW264.7, inhibit the cell proliferation, and promote the cell apoptosis. After ox-LDL induction, expression of Caspase-3 in macrophages RAW264.7 was up-regulated, and that of glucose regulated protein 78 was down-regulated. The transfection of miRNA-9 mimics could greatly inhibit the expression of PDGFR-ß mRNA and proteins in the cells. In addition, the results of the dual-luciferase reporter gene showed that the ratio of luciferase activity was significantly reduced after the miRNA-9 mimic and the wild-type PDGFR-ß plasmid were co-transfected. In summary, ox-LDL could induce foaming of macrophages and promote cell apoptosis, and miRNA-9 could target and bind to the 3'UTR region of PDGFR-ß, thereby inhibiting the gene expression.


Subject(s)
Apoptosis , Lipoproteins, LDL/pharmacology , Macrophages/metabolism , MicroRNAs/metabolism , Receptors, Platelet-Derived Growth Factor/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , Caspase 3/metabolism , Cell Proliferation/drug effects , Endoplasmic Reticulum Chaperone BiP/metabolism , Genes, Reporter , Luciferases/metabolism , Macrophages/drug effects , Macrophages/pathology , Mice , MicroRNAs/genetics , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism
8.
J Cell Mol Med ; 25(20): 9851-9862, 2021 10.
Article in English | MEDLINE | ID: mdl-34523794

ABSTRACT

Adiponectin is an adipocyte-derived hormone, which is closely associated with the development of Alzheimer's disease (AD) and has potential preventive and therapeutic significance. In the present study, we explored the relationship between adiponectin and circadian rhythm disorder in AD, the effect of adiponectin on the abnormal expression of Bmal1 mRNA/protein induced by amyloid-ß protein 31-35 (Aß31-35), and the underlying mechanism of action. We found that adiponectin-knockout mice exhibited amyloid-ß deposition, circadian rhythm disorders and abnormal expression of Bmal1. Adiponectin ameliorated the abnormal expression of the Bmal1 mRNA/protein caused by Aß31-35 by inhibiting the activity of glycogen synthase kinase 3ß (GSK3ß). These results suggest that adiponectin deficiency could induce circadian rhythm disorders and abnormal expression of the Bmal1 mRNA/protein, whilst exogenous administration of adiponectin may improve Aß31-35-induced abnormal expression of Bmal1 by inhibiting the activity of GSK3ß, thus providing a novel idea for the treatment of AD.


Subject(s)
Adiponectin/metabolism , Amyloid beta-Peptides/metabolism , Chronobiology Disorders/etiology , Chronobiology Disorders/metabolism , Peptide Fragments/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Adiponectin/genetics , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/adverse effects , Animals , Cell Line , Chronobiology Disorders/pathology , Disease Models, Animal , Disease Susceptibility , Gene Expression , Glycogen Synthase Kinase 3 beta/metabolism , Male , Mice , Mice, Knockout , Peptide Fragments/adverse effects , Protein Aggregation, Pathological/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/pathology
9.
J Immunol Res ; 2021: 1815098, 2021.
Article in English | MEDLINE | ID: mdl-34307691

ABSTRACT

Adiponectin is a small peptide secreted and a key component of the endocrine system and immune system. Although globular adiponectin protects myocardial ischemia/reperfusion-induced cardiomyocyte injury, the protective mechanisms remain largely unresolved. Using a neonatal rat ventricular myocyte hypoxia/reoxygenation model, we investigated the role of its potential mechanisms of necroptosis in globular adiponectin-mediated protection in hypoxia/reoxygenation-induced cardiomyocyte injury as compared to apoptosis. We found that globular adiponectin treatment attenuated cardiomyocyte injury as indicated by increased cell viability and reduced lactate dehydrogenase release following hypoxia/reoxygenation. Immunofluorescence staining and Western blotting demonstrated that both necroptosis and apoptosis were triggered by hypoxia/reoxygenation and diminished by globular adiponectin. Necrostatin-1 (RIP1-specific inhibitor) and Z-VAD-FMK (pan-caspase inhibitor) only mimicked the inhibition of necroptosis and apoptosis, respectively, by globular adiponectin in hypoxia/reoxygenation-treated cardiomyocytes. Globular adiponectin attenuated reactive oxygen species production, oxidative damage, and p38MAPK and NF-κB signaling, all important for necroptosis and apoptosis. Collectively, our study suggests that globular adiponectin inhibits hypoxia/reoxygenation-induced necroptosis and apoptosis in cardiomyocytes probably by reducing oxidative stress and interrupting p38MAPK signaling.


Subject(s)
Adiponectin/metabolism , Myocardial Reperfusion Injury/immunology , Myocytes, Cardiac/pathology , Animals , Animals, Newborn , Apoptosis/immunology , Cell Hypoxia/immunology , Cell Survival , Cells, Cultured , Culture Media/metabolism , Disease Models, Animal , Female , Humans , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/immunology , Necroptosis/immunology , Oxidative Stress/immunology , Pregnancy , Primary Cell Culture , Rats , Reactive Oxygen Species/metabolism
10.
J Cell Mol Med ; 25(17): 8464-8478, 2021 09.
Article in English | MEDLINE | ID: mdl-34322993

ABSTRACT

Cardiomyocytes autophagy is essential for maintaining cardiac function. Our previous studies have found that ß1 -adrenergic receptor autoantibody (ß1 -AA) induced the decreased myocardial autophagic flux, which resulted in cardiomyocyte death and cardiac dysfunction. And other studies demonstrated that ß1 -AA induced the decrease of AMPK phosphorylation, the key hub of autophagy pathway, while adiponectin up-regulated autophagic flux mediated by AMPK. However, it is not clear whether adiponectin improves the inhibition of myocardial autophagic flux induced by ß1 -AA by up-regulating the level of AMPK phosphorylation. In this study, it has been confirmed that ß1 -AA induced the decrease of AMPK phosphorylation level in both vivo and vitro. Moreover, pretreatment of cardiomyocytes with AMPK inhibitor Compound C could further reduce the autophagic flux induced by ß1 -AA. Adiponectin deficiency could aggravate the decrease of myocardial AMPK phosphorylation level, autophagic flux and cardiac function induced by ß1 -AA. Further, exogenous adiponectin could reverse the decline of AMPK phosphorylation level and autophagic flux induced by ß1 -AA and even reduce cardiomyocyte death. While pretreated with the Compound C, the adiponectin treatment did not improve the decreased autophagosome formation, but still improved the decreased autophagosome clearance induced by ß1 -AA in cardiomyocytes. This study is the first time to confirm that ß1 -AA could inhibit myocardial autophagic flux by down-regulating AMPK phosphorylation level. Adiponectin could improve the inhibition of myocardial autophagic flux induced by ß1 -AA partly dependent on AMPK, so as to provide an experimental basis for the treatment of patients with ß1 -AA-positive cardiac dysfunction.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adiponectin/metabolism , Heart Diseases/metabolism , Receptors, Adrenergic, beta-1/metabolism , Animals , Autophagy , Cell Line , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoblasts , Rats
12.
Hum Cell ; 34(1): 49-59, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32959354

ABSTRACT

ATF3 (activating transcription factor 3) is a member of the mammalian activation transcription factor/cAMP-responsive element-binding (CREB) family. It plays a role in inflammation and innate immunity, and suggests that ATF3 is associated with atherosclerosis. In our study, we analyzed datasets of atherosclerosis from the NCBI-GEO (Gene Expression Omnibus) database and found that expression levels of ATF3 were lower in macrophages from ruptured atherosclerotic plaques than from stable atherosclerotic plaques. Expression levels of ATF3 correlated with the stability of atherosclerotic plaques. KEGG analysis of different expression genes (DEGs) between ruptured and stable atherosclerotic plaques was performed by Metascape database. The PI3K-AKT pathway may be a potential pathway of the formation of ruptured atherosclerotic plaques. High-fat diet-induced atherosclerosis apoE-/- mice were divided into two groups: a model group and an ATF3 overexpression (OE)-group. Tests on atherosclerotic plaques in the aortic root suggested that absence of ATF3 and increase of macrophages may be risk factors for the formation of ruptured atherosclerotic plaques. We found decreased areas of lesions in aortic roots and branches of aortic arch, as well as increased lesional content of macrophages as well as TUNEL-positive areas. Consistent with these results, we found reduced degradation and incidence of elastic plate cracks accompanied by suppressed MMPs expression and transduction pathway protein PI3K/AKT activation. These data suggest that ATF3 is a signaling molecule that mediates the progression and stability of atherosclerotic plaques. ATF3 could be a potential new biomarker for the prognosis of atherosclerosis and may be a therapeutic target to reduce atherosclerosis.


Subject(s)
Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/physiology , Atherosclerosis/diagnosis , Atherosclerosis/genetics , Plaque, Atherosclerotic/genetics , Activating Transcription Factor 3/metabolism , Animals , Atherosclerosis/immunology , Atherosclerosis/therapy , Biomarkers , Gene Expression , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases/metabolism , Plaque, Atherosclerotic/etiology , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
13.
Mol Med Rep ; 23(2)2021 02.
Article in English | MEDLINE | ID: mdl-33300086

ABSTRACT

Intermedin (IMD) is a calcitonin/calcitonin­related peptide that elicits cardioprotective effects in a variety of heart diseases, such as cardiac hypertrophy and heart failure. However, the molecular mechanism of IMD remains unclear. The present study investigated the effects of IMD on neonatal rat ventricular myocytes treated with thapsigargin. The results of the present study demonstrated that thapsigargin induced apoptosis in cardiomyocytes in a dose­ and time­dependent manner. Thapsigargin induced endoplasmic reticulum stress, as determined by increased expression levels of 78­kDa glucose­regulated protein, C/EBP­homologous protein and caspase­12, which were dose­dependently attenuated by pretreatment with IMD. In addition, IMD treatment counteracted the thapsigargin­induced suppression of sarco/endoplasmic reticulum Ca2+­ATPase (SERCA) activity and protein expression levels, and cytoplasmic Ca2+ overload. IMD treatment also augmented the phosphorylation of phospholamban, which is a crucial regulator of SERCA. Additionally, treatment with the protein kinase A antagonist H­89 inhibited the IMD­mediated cardioprotective effects, including SERCA activity restoration, anti­Ca2+ overload, endoplasmic reticulum stress inhibition and antiapoptosis effects. In conclusion, the results of the present study suggested that IMD may protect cardiomyocytes against thapsigargin­induced endoplasmic reticulum stress and the associated apoptosis at least partly by activating the protein kinase A/SERCA pathway.


Subject(s)
Adrenomedullin/metabolism , Calcium Signaling/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Endoplasmic Reticulum Stress/drug effects , Myocytes, Cardiac/metabolism , Neuropeptides/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thapsigargin/pharmacology , Adrenomedullin/genetics , Animals , Calcium Signaling/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Endoplasmic Reticulum Stress/genetics , Female , Male , Neuropeptides/genetics , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
14.
Cell Chem Biol ; 27(12): 1483-1499.e9, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33186540

ABSTRACT

H2S-producing enzymes in bacteria have been shown to be closely engaged in the process of microbial survival and antibiotic resistance. However, no inhibitors have been discovered for these enzymes, e.g., 3-mercaptopyruvate sulfurtransferase (MST). In the present study, we identified several classes of inhibitors for Escherichia coli MST (eMST) through high-throughput screening of ∼26,000 compounds. The thiazolidinedione-type inhibitors were found to selectively bind to Arg178 and Ser239 residues of eMST but hardly affected human MST. Moreover, the pioglitazone of this class concentration dependently accumulates the 3-mercaptopyruvate substrate and suppresses the H2S and reactive sulfane sulfur products in bacteria. Importantly, pioglitazone could potentiate the level of reactive oxygen species in cellulo and consequently enhance the antimicrobial effects of gentamicin and macrophages in culture. This study has identified the bioactive inhibitor of eMST, paving the way for the pharmacological targeting of eMST to synergistically control the survival of E. coli.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Escherichia coli/enzymology , Sulfurtransferases/antagonists & inhibitors , Drug Evaluation, Preclinical , Drug Synergism , Escherichia coli/physiology , High-Throughput Screening Assays , Humans
15.
Med Sci Monit ; 26: e924016, 2020 Aug 09.
Article in English | MEDLINE | ID: mdl-32772038

ABSTRACT

BACKGROUND The objective of the study was to explore the role of long non-coding RNA SNHG8 (lncRNA SNHG8) in myocardial infarction (MI) and the related mechanism of action. MATERIAL AND METHODS In vitro model of MI was established by hypoxia induction in cardiomyocyte line H9c2 cells. H9c2 cells were transfected with control-plasmid, SNHG8-plasmid, control-shRNA and SNHG8-shRNA. Quantitative real-time polymerase chain reaction (qRT-PCR) assay was performed to measure transfection efficiency. Creatine kinase-muscle/brain (CK-MB) release, cardiac troponin 1 (cTnI) release and mitochondria viability were detected by using related detection kits. MTT (3-(45)-dimethylthiahiazo (-z-y 1)-35-diphenytetrazoliumromide) assay was used to detect cell viability and flow cytometry analysis was used to detect cell apoptosis. Western blot assay was performed to measure protein expression of cleaved-Caspase3, p-p65 and p65. Enzyme-linked immunosorbent assay (ELISA) and qRT-PCR assay were performed to detect expression of interleukin (IL)-1ß, tumor necrosis factor (TNF)-alpha and IL-6. RESULTS LncRNA SNHG8 was overexpressed in hypoxia-induced cardiomyocytes. SNHG8-plasmid increased lncRNA SNHG8 expression, CK-MB release, cTnI release, and mitochondria viability in hypoxia-induced H9c2 cells. In addition, SNHG8-plasmid reduced cell viability, induced cell apoptosis, and increased expression of cleaved-caspase3, IL-1ß, TNF-alpha, IL-6, and p-p65 in hypoxia-induced H9c2 cells, while the effects of SNHG8-shRNA were opposite. CONCLUSIONS We demonstrated that lncRNA SNHG8 affected myocardial infarction by affecting hypoxia-induced cardiomyocyte injury via regulation of the NF-kappaB pathway.


Subject(s)
Cell Hypoxia/genetics , DNA Damage/genetics , Myocardial Infarction/genetics , Myocytes, Cardiac/pathology , RNA, Long Noncoding/physiology , Animals , Apoptosis , Biomarkers/metabolism , Cell Line , Cell Survival , Gene Knockdown Techniques , Inflammation Mediators/metabolism , Myocardial Infarction/metabolism , Rats
16.
Biochem Cell Biol ; 98(5): 583-590, 2020 10.
Article in English | MEDLINE | ID: mdl-32413267

ABSTRACT

Research has shown that some circular RNAs (circRNA) are abnormally expressed in the process of myocardial fibrosis, but the mechanism behind this was unknown. In the process of inducing cardiac fibroblast (CF) activation with TGF-ß1 or Ang II, the expression of circRNA circ_BMP2K and miR-455-3p were significantly inhibited, whereas the expression of SUMO1 was promoted. The results from our dual luciferase reporter gene assays, RIP assays, and pull-down assays show that miR-455-3p directly binds circ_BMP2K, thereby mutually promoting their expression levels. SUMO1 is a target gene of miR-455-3p, and circ_BMP2K enhances the inhibitory effects of miR-455-3p on the expression of SUMO1. Further study showed that both circ_BMP2K and miR-455-3p inhibited the expression of alpha-SMA as well as type I and type III collagen, whereas SUMO1 promoted their expression, and miR-455-3p inhibitors or overexpression of SUMO1 reversed the effects of circ_BMP2K and miR-455-3p. Circ_BMP2K and miR-455-3p inhibits cell proliferation and migration and promotes the apoptosis of CFs, but SUMO1 has the opposite effects; miR-455-3p inhibitors or overexpression of SUMO1 reverses the effects of circ_BMP2K and miR-455-3p. Thus, circ_BMP2K promotes the expression of miR-455-3p, down-regulates the expression of SUMO1, and finally, inhibits the activation, growth, and migration of CFs. These results could provide important therapeutic targets and a theoretical basis for regulating the process of myocardial fibrosis.


Subject(s)
Fibroblasts/metabolism , MicroRNAs/metabolism , RNA, Circular/metabolism , SUMO-1 Protein/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Fibroblasts/pathology , Humans , MicroRNAs/genetics , RNA, Circular/genetics , SUMO-1 Protein/genetics
17.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(9): 929-938, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28602962

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) plays a crucial role in reverse cholesterol transport and exhibits anti-atherosclerosis effects. Some microRNAs (miRs) regulate ABCA1 expression, and recent studies have shown that miR-20a/b might play a critical role in atherosclerotic diseases. Here, we attempted to clarify the potential contribution of miR-20a/b in post-transcriptional regulation of ABCA1, cholesterol efflux, and atherosclerosis. We performed bioinformatics analysis and found that miR-20a/b was highly conserved and directly bound to ABCA1 mRNA with low binding free energy. Luciferase-reporter assay also confirmed that miR-20a/b significantly reduced luciferase activity associated with the ABCA1 3' untranslated region reporter construct. Additionally, miR-20a/b decreased ABCA1 expression, which, in turn, decreased cholesterol efflux and increased cholesterol content in THP-1 and RAW 264.7 macrophage-derived foam cells. In contrast, miR-20a/b inhibitors increased ABCA1 expression and cholesterol efflux, decreased cholesterol content, and inhibited foam-cell formation. Consistent with our in vitro results, miR-20a/b-treated ApoE-/- mice showed decreased ABCA1expression in the liver and reductions of reverse cholesterol transport in vivo. Furthermore, miR-20a/b regulated the formation of nascent high-density lipoprotein and promoted atherosclerotic development, whereas miR-20a/b knockdown attenuated atherosclerotic formation. miR-20 is a new miRNA capable of targeting ABCA1 and regulating ABCA1 expression. Therefore, miR-20 inhibition constitutes a new strategy for ABCA1-based treatment of atherosclerosis.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Cholesterol/metabolism , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional/physiology , 3' Untranslated Regions , Animals , Atherosclerosis/metabolism , Cell Line , Foam Cells/metabolism , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Lipoproteins, HDL/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells
18.
Exp Ther Med ; 13(6): 2757-2762, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28587337

ABSTRACT

The objective of the present study was to investigate the effect of adiponectin (APN) on macrophage reverse cholesterol transport (RCT) in adiponectin-/- knockout mice (APN-/-mice) and its possible anti-atherosclerotic mechanism. A total of 30 male APN-/-mice were randomly divided into the control group and four intervention groups. The intervention groups were treated with intraperitoneal injections of APN, at doses of 50, 150, 200 and 250 µg/(kg/day), respectively, for 4 weeks. The control group received normal saline. After 4 weeks, serum lipid levels were measured, the degree of severity of atherosclerotic lesions was observed by light microscopy, the 3H-TC (APN-/-mice treated with intraperitoneal injections of 3H-TC-labeled macrophages) radioactivity in serum, liver, and feces, and the expression of ABCA1 mRNA and protein in liver were determined. Compared with the control group, serum triglycerides, total cholesterol, and low-density lipoproteins levels in the intervention groups were significantly decreased, while high-density lipoprotein was increased. The severity of aortic atherosclerotic lesions in the intervention groups was milder than in the control group, which had obvious aortic atherosclerotic lesions, large lipid deposition on vessel walls, and the formation of atheromatous plaques. In the intervention groups, serum 3H-TC content was significantly decreased (P<0.05), but the 3H-TC content in liver and feces was significantly increased (P<0.05). The levels of ABCA1 mRNA in liver of the intervention groups were significantly increased in a dose-dependent manner. In conclusion, APN can promote RCT and intracellular cholesterol efflux by upregulating the expression of ABCA1, to delay the occurrence and development of atherosclerosis.

19.
Cell Physiol Biochem ; 38(5): 1906-14, 2016.
Article in English | MEDLINE | ID: mdl-27160732

ABSTRACT

BACKGROUND/AIMS: To detect the changes of high density lipoprotein (HDL) and its subtypes in serum of patients with coronary heart disease (CHD). METHODS: 337 hospitalized patients were selected from our hospital during August, 2014 - January, 2015, and divided into CHD group (n = 190) and control group (n = 127). Lipoprint lipoprotein analyzer was used to classify low density lipoprotein (LDL) particle size and its sub-components, as well as HDL particle size and its sub-components. The changes of the subtypes in patients with CHD were statistically analyzed. The possible mechanism was explored. RESULTS: (1) Compared with the control group, the concentration of HDL in CHD patients reduced, HDLL significantly decreased (P < 0.001), while HDLS increased (P < 0.001); (2) In the patients with HDL less than 1.04 mmol/L among CHD, all HDL subtypes reduced, but HDLL had the most significant decreased; (3) HDL and all HDL subtypes were positively correlated with apolipoprotein A-I (apoA-I), of which, HDLL had the biggest correlation with apoA-I (P < 0.001); (4) HDL subtypes had good correlation with HDL, of which, HDLM had a maximum correlation with HDL (P < 0.001). CONCLUSION: HDL maturation disorders existed in the serum of CHD patients, HDLL may be protected factor for CHD, whose decrease was closely related wit the risk increase of CHD. The cardiovascular protection function of HDLL may be related with apoA-I content.


Subject(s)
Coronary Artery Disease/pathology , Aged , Apolipoprotein A-I/blood , Body Mass Index , Case-Control Studies , Coronary Artery Disease/blood , Female , Humans , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Male , Middle Aged , Particle Size , Triglycerides/blood
20.
Mol Med Rep ; 13(6): 5021-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27121797

ABSTRACT

Platelet activation is important in hypertension­induced cardiac inflammation and fibrosis. P-selectin expression significantly (P<0.05) increases when platelets are activated during hypertension. Although P­selectin recruits leukocytes to sites of inflammation, the role of P­selectin in cardiac inflammation and fibrosis remains to be elucidated. The present study aimed to investigate whether platelet­derived P­selectin promotes hypertensive cardiac inflammation and fibrosis. P­selectin knockout (P­sel KO) mice and wild­type (WT) C57BL/6 littermates were infused with angiotensin II (Ang II) at 1,500 ng/kg/min for 7 days and then cross­transplanted with platelets originating from either WT or P­sel KO mice. P­selectin expression was increased in the myocardium and plasma of hypertensive mice, and the P­sel KO mice exhibited significantly (P<0.05) reduced cardiac fibrosis. The fibrotic areas were markedly smaller in the hearts of P­sel KO mice compared with WT mice, as assessed by Masson's trichrome staining. In addition, α­smooth muscle actin and transforming growth factor ß1 (TGF­ß1) expression levels were decreased in the P­sel KO mice, as assessed by immunohistochemistry. Following platelet transplantation into P­sel KO mice, the number of Mac­2 (galectin­3)­ and TGF­ß1­positive cells was increased in mice that received WT platelets compared with those that received P­sel KO platelets, and the mRNA expression levels of collagen I and TGF­ß1 were also increased. The results from the present study suggest that activated platelets secrete P­selectin to promote cardiac inflammation and fibrosis in Ang II­induced hypertension.


Subject(s)
Angiotensin II/metabolism , Blood Platelets/metabolism , Myocarditis/etiology , Myocarditis/metabolism , P-Selectin/metabolism , Platelet Activation , Angiotensin II/blood , Animals , Blood Platelets/drug effects , Disease Models, Animal , Endothelial Cells/metabolism , Fibrosis , Gene Expression , Hypertension/chemically induced , Hypertension/complications , Immunohistochemistry , Male , Mice , Mice, Knockout , Myocarditis/diagnosis , Myocardium/metabolism , Myocardium/pathology , P-Selectin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...