Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Clin Immunol ; 255: 109767, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37689092

ABSTRACT

RATIONALE: A persistent autoimmune and inflammatory response plays a critical role in the progression of atherosclerosis. The transcription factor forkhead box P3 (Foxp3)+CD4+ regulatory T cells (Foxp3+ Tregs) attenuate atherosclerosis. Latency-associated peptide (LAP)+CD4+ T cells are a new class of Tregs whose role in atherosclerosis is unknown. OBJECTIVE: To investigate the function of CD4+LAP+ Tregs in inhibiting inflammation and preventing atherosclerosis. METHODS AND RESULTS: Depletion of CD4+LAP+ Tregs results in aggravated inflammation and atherosclerotic lesions. Mechanistically, CD4+LAP+ Treg depletion was associated with decreased M2-like macrophages and increased Th1 and Th17 cells, characterized by increased unstable plaque promotion and decreased expression of inflammation-resolving factors in both arteries and immune organs. In contrast, adoptive transfer of CD4+LAP+ Tregs to ApoE-/- mice or CD4-/-ApoE-/- mice led to decreased atherosclerotic lesions. Compared with control animals, adoptive transfer of CD4+LAP+ Tregs induced M2-like macrophage differentiation within the atherosclerotic lesion and spleen, associated with increased collagen and α-SMA in plaques and decreased expression of MMP-2 and MMP-9. Mechanistic studies reveal that isolated CD4+LAP+ Tregs exhibit a tolerance phenotype, with increased expression of inhibitory cytokines and coinhibitory molecules. After coculture with CD4+LAP+ Tregs, monocytes/macrophages display typical features of M2 macrophages, including upregulated expression of CD206 and Arg-1 and decreased production of MCP-1, IL-6, IL-1ß and TNF-α, which was almost abrogated by transwell and partially TGF-ß1 neutralization. RNA-seq analysis showed different gene expression profiles between CD4+LAP+ Tregs and LAP-CD4+ T cells and between CD4+LAP+ Tregs of ApoE-/- mice and CD4+LAP+ Tregs of C57BL/6 mice, of which Fancd2 and IL4i1 may contribute to the powerful inhibitory properties of CD4+LAP+ Tregs. Furthermore, the number and the suppressive properties of CD4+LAP+ Tregs were impaired by oxLDL. CONCLUSIONS: Our data indicate that the remaining CD4+LAP+ Tregs play a protective role in atherosclerosis by modulating monocyte/macrophage differentiation and regulatory factors, which may partly explain the protective effect of T cells tolerance in atherosclerosis. Moreover, adoptive transfer of CD4+LAP+ Tregs constitutes a novel approach to treat atherosclerosis.

2.
Mediators Inflamm ; 2022: 2054879, 2022.
Article in English | MEDLINE | ID: mdl-36405994

ABSTRACT

Atherosclerosis, which is characterized by chronic inflammation in the arterial wall, is driven by immune cells and cytokines. Recent evidence indicated that interleukin (IL)-27 showed pleiotropic properties in immune diseases. However, precise mechanisms of IL-27, especially in atherosclerosis remains unknown. In our research, we examined the influence of the administration of IL-27 and an anti-IL-27p28 antibody (anti-IL-27p28-Ab) on both the initiation and the progression of atherosclerosis. In the groups (both the initiation and the progression) receiving recombinant IL-27 administration, the formation of atherosclerotic plaques was suspended, and the percentage of regulatory T cells (LAP+ or Foxp3+) in the spleen and peripheral blood was increased. Meanwhile, the number of T helper 1 (Th1) and T helper 17 (Th17) cells was decreased. In the peripheral blood plasma, TGF-ß and IL-10 expression were increased, while the levels of IFN-γ and IL-17 were reduced. As for lesions, the mRNA expression of Foxp3, TGF-ß, and IL-10 was increased, while that of IFN-γ and IL-17 was reduced. In the anti-IL-27p28 antibody groups, we obtained opposite results. We also observed that DCs treated with IL-27 display a tolerogenic phenotype and that IL-27-treated tolerogenic DCs (tDCs) are likely to play a protective role during atherosclerosis. Our study indicates that IL-27 or adoptive transfer of IL-27 loaded tDCs may be a new therapeutic approach in atherosclerosis.


Subject(s)
Atherosclerosis , Interleukin-27 , Mice , Animals , T-Lymphocytes, Regulatory/metabolism , Interleukin-10/metabolism , Interleukin-27/metabolism , Interleukin-17/metabolism , Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Interleukins/metabolism , Transforming Growth Factor beta/metabolism , Immunologic Factors/therapeutic use , Forkhead Transcription Factors/metabolism , Dendritic Cells/metabolism
3.
J Cell Mol Med ; 24(1): 371-384, 2020 01.
Article in English | MEDLINE | ID: mdl-31746138

ABSTRACT

Excessive immune-mediated inflammatory reaction plays a deleterious role in ventricular remodelling after myocardial infarction (MI). Interleukin (IL)-38 is a newly characterized cytokine of the IL-1 family and has been reported to exert a protective effect in some autoimmune diseases. However, its role in cardiac remodelling post-MI remains unknown. In this study, we found that the expression of IL-38 was increased in infarcted heart after MI induced in C57BL/6 mice by permanent ligation of the left anterior descending artery. In addition, our data showed that ventricular remodelling after MI was significantly ameliorated after recombinant IL-38 injection in mice. This amelioration was demonstrated by better cardiac function, restricted inflammatory response, attenuated myocardial injury and decreased myocardial fibrosis. Our results in vitro revealed that IL-38 affects the phenotype of dendritic cells (DCs) and IL-38 plus troponin I (TNI)-treated tolerogenic DCs dampened adaptive immune response when co-cultured with CD4+ T cells. In conclusion, IL-38 plays a protective effect in ventricular remodelling post-MI, one possibility by influencing DCs to attenuate inflammatory response. Therefore, targeting IL-38 may hold a new therapeutic potential in treating MI.


Subject(s)
Interleukin-1/metabolism , Myocardial Infarction/physiopathology , Ventricular Remodeling , Animals , Apoptosis/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Fibrosis , Immune Tolerance/drug effects , Inflammation/pathology , Interleukin-1/genetics , Interleukin-1/pharmacology , Male , Mice, Inbred C57BL , Myocardial Infarction/genetics , Myocardial Infarction/immunology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Troponin I/metabolism , Ventricular Remodeling/drug effects
4.
Circ Res ; 120(11): 1754-1767, 2017 May 26.
Article in English | MEDLINE | ID: mdl-28348007

ABSTRACT

RATIONALE: Acute coronary syndrome (ACS) is a leading cause of death worldwide. Immune functions play a vital role in ACS development; however, whether epigenetic modulation contributes to the regulation of blood immune cells in this disease has not been investigated. OBJECTIVE: We conducted an epigenome-wide analysis with circulating immune cells to identify differentially methylated genes in ACS. METHODS AND RESULTS: We examined genome-wide methylation of whole blood in 102 ACS patients and 101 controls using HumanMethylation450 array, and externally replicated significant discoveries in 100 patients and 102 controls. For the replicated loci, we further analyzed their association with ACS in 6 purified leukocyte subsets, their correlation with the expressions of annotated genes, and their association with cardiovascular traits/risk factors. We found novel and reproducible association of ACS with blood methylation at 47 cytosine-phosphoguanine sites (discovery: false discovery rate <0.005; replication: Bonferroni corrected P<0.05). The association of methylation levels at these cytosine-phosphoguanine sites with ACS was further validated in at least 1 of the 6 leukocyte subsets, with predominant contributions from CD8+ T cells, CD4+ T cells, and B cells. Blood methylation of 26 replicated cytosine-phosphoguanine sites showed significant correlation with expressions of annotated genes (including IL6R, FASLG, and CCL18; P<5.9×10-4), and differential gene expression in case versus controls corroborated the observed differential methylation. The replicated loci suggested a role in ACS-relevant functions including chemotaxis, coronary thrombosis, and T-cell-mediated cytotoxicity. Functional analysis using the top ACS-associated methylation loci in purified T and B cells revealed vital pathways related to atherogenic signaling and adaptive immune response. Furthermore, we observed a significant enrichment of the replicated cytosine-phosphoguanine sites associated with smoking and low-density lipoprotein cholesterol (Penrichment≤1×10-5). CONCLUSIONS: Our study identified novel blood methylation alterations associated with ACS and provided potential clinical biomarkers and therapeutic targets. Our results may suggest that immune signaling and cellular functions might be regulated at an epigenetic level in ACS.


Subject(s)
Acute Coronary Syndrome/blood , Acute Coronary Syndrome/genetics , DNA Methylation/physiology , Epigenesis, Genetic/physiology , Genome-Wide Association Study/methods , Acute Coronary Syndrome/epidemiology , Aged , Case-Control Studies , China/epidemiology , Female , Humans , Male , Middle Aged
5.
Mediators Inflamm ; 2019: 9515346, 2019.
Article in English | MEDLINE | ID: mdl-31686988

ABSTRACT

BACKGROUND: Interleukin-37 (IL-37) acts as an inhibitor of innate and adaptive immunity. However, the exact role of IL-37 in the patients with acute coronary syndrome (ACS) remains to be elucidated. METHODS: Patients were classified into 4 groups: normal coronary artery (NCA), stable angina (SA), unstable angina (UA), and acute myocardial infarction (AMI). The circulating Treg, Th1, and Th17 frequencies were measured. The effect of IL-37 on stimulated peripheral blood mononuclear cells (PBMCs) and the influence of IL-37 on DCs were explored. In addition, the role of IL-37-treated tDCs on Treg cell expansion and the stability of these tDCs were also tested. RESULTS: Our results showed that the circulating Treg frequencies were decreased, while Th1 and Th17 frequencies were increased in ACS patients, and that IL-37 expanded Tregs but suppressed Th1 and Th17 cells in activated PBMCs derived from ACS patients. Of note, IL-37-treated human DCs obtained a tolerogenic phenotype, and such tDCs promoted expansion of Tregs and decreased the Th1 and Th17 populations when cocultured with CD4+ T cells. Interestingly, IL-37-treated DCs from patients with ACS are phenotypically and functionally comparable to IL-37-treated DCs from NCA patients, and tolerogenic properties of IL-37-treated DCs were highly stable. CONCLUSION: In conclusion, our results reveal a beneficial role of IL-37 in the patients with ACS and suggest that autologous IL-37-treated tDCs may be a novel therapeutic strategy for the patients with ACS.


Subject(s)
Acute Coronary Syndrome/metabolism , Interleukin-1/pharmacology , Angina, Stable/metabolism , Angina, Unstable/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Flow Cytometry , Humans , Interleukin-17/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism
6.
J Cell Biochem ; 119(9): 7212-7217, 2018 09.
Article in English | MEDLINE | ID: mdl-29932232

ABSTRACT

Thymoquinone (TQ), as the active constituents of black cumin (Nigella sativa) seed oil, has been reported to have potential protective effects on the cardiovascular system. This study aimed to investigate the effects and the underlying mechanisms of TQ on myocardial ischemia-reperfusion (I/R) injury in Langendorff-perfused rat hearts. Wister rat hearts were subjected to I/R and the experimental group were pretreated with TQ prior to I/R. Hemodynamic parameters, myocardial infarct size, cardiac marker enzymes, superoxide dismutase (SOD), malondialdehyde (MDA) content, and cardiomyocyte apoptosis were assayed. Compared with the untreated group, TQ preconditioning significantly improved cardiac function, reduced infarct size, decreased cardiac lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) levels, suppressed enedoxidative stress, and apoptosis. In addition, TQ treatment promoted autophagy, which was partially reversed by chloroquine (CQ), a kind of autophagy blocker. Our study suggests that TQ can protect heart against I/R injury, which is associated with anti-oxidative and anti-apoptotic effects through activation of autophagy.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Benzoquinones/therapeutic use , Cardiotonic Agents/therapeutic use , Heart/drug effects , Myocardial Reperfusion Injury/drug therapy , Analysis of Variance , Animals , Creatine Kinase, MB Form/antagonists & inhibitors , Heart Rate/drug effects , Isolated Heart Preparation/methods , L-Lactate Dehydrogenase/antagonists & inhibitors , Male , Myocardial Infarction/physiopathology , Oxidative Stress/drug effects , Plant Oils/chemistry , Rats , Rats, Wistar , Ventricular Pressure/drug effects
7.
Eur Heart J ; 38(46): 3449-3460, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29029073

ABSTRACT

AIMS: In this study, we aimed to clinically and genetically characterize LVNC patients and investigate the prevalence of variants in known and novel LVNC disease genes. INTRODUCTION: Left ventricular non-compaction cardiomyopathy (LVNC) is an increasingly recognized cause of heart failure, arrhythmia, thromboembolism, and sudden cardiac death. We sought here to dissect its genetic causes, phenotypic presentation and outcome. METHODS AND RESULTS: In our registry with follow-up of in the median 61 months, we analysed 95 LVNC patients (68 unrelated index patients and 27 affected relatives; definite familial LVNC = 23.5%) by cardiac phenotyping, molecular biomarkers and exome sequencing. Cardiovascular events were significantly more frequent in LVNC patients compared with an age-matched group of patients with non-ischaemic dilated cardiomyopathy (hazard ratio = 2.481, P = 0.002). Stringent genetic classification according to ACMG guidelines revealed that TTN, LMNA, and MYBPC3 are the most prevalent disease genes (13 patients are carrying a pathogenic truncating TTN variant, odds ratio = 40.7, Confidence interval = 21.6-76.6, P < 0.0001, percent spliced in 76-100%). We also identified novel candidate genes for LVNC. For RBM20, we were able to perform detailed familial, molecular and functional studies. We show that the novel variant p.R634L in the RS domain of RBM20 co-segregates with LVNC, leading to titin mis-splicing as revealed by RNA sequencing of heart tissue in mutation carriers, protein analysis, and functional splice-reporter assays. CONCLUSION: Our data demonstrate that the clinical course of symptomatic LVNC can be severe. The identified pathogenic variants and distribution of disease genes-a titin-related pathomechanism is found in every fourth patient-should be considered in genetic counselling of patients. Pathogenic variants in the nuclear proteins Lamin A/C and RBM20 were associated with worse outcome.


Subject(s)
Hypertrophy, Left Ventricular/genetics , Mutation/genetics , Adult , Arrhythmias, Cardiac/genetics , Cardiomyopathy, Dilated/genetics , Connectin/genetics , Death, Sudden, Cardiac/etiology , Female , Genetic Predisposition to Disease/genetics , Humans , Lamin Type A/genetics , Male , Pedigree , RNA-Binding Proteins/genetics
8.
Immunology ; 151(3): 291-303, 2017 07.
Article in English | MEDLINE | ID: mdl-28207945

ABSTRACT

Dilated cardiomyopathy (DCM) is a lethal inflammatory heart disease and closely connected with dysfunction of the immune system. Glycoprotein A repetitions predominant (GARP) expressed on activated CD4+ T cells with suppressive activity has been established. This study aimed to investigate the frequency and function of circulating CD4+  CD25+  GARP+ regulatory T (Treg) cells in DCM. Forty-five DCM patients and 46 controls were enrolled in this study. There was a significant increase in peripheral T helper type 1 (Th1) and Th17 number and their related cytokines [interferon-γ (IFN-γ), interleukin (IL-17)], and an obvious decrease in Treg number, transforming growth factor-ß1 (TGF-ß1 ) levels and the expression of forkhead box P3 (FOXP3) and GARP in patients with DCM compared with controls. In addition, the suppressive function of CD4+  CD25+  GARP+ Treg cells was impaired in DCM patients upon T-cell receptor stimulation detected using CFSE dye. Lower level of TGF-ß1 and higher levels of IFN-γ and IL-17 detected using ELISA were found in supernatants of the cultured CD4+  CD25+  GARP+ Treg cells in DCM patients compared with controls. Together, our results indicate that CD4+  CD25+  GARP+ Treg cells are defective in DCM patients and GARP seems to be a better molecular definition of the regulatory phenotype. Therefore, it might be an attractive stategy to pay more attention to GARP in DCM patients.


Subject(s)
Adaptive Immunity , Cardiomyopathy, Dilated/immunology , Cell Proliferation , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Activation , Membrane Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Adult , CD4 Lymphocyte Count , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/metabolism , Case-Control Studies , Cell Communication , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Female , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Membrane Proteins/metabolism , Middle Aged , Phenotype , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism
9.
Cell Physiol Biochem ; 44(5): 1810-1827, 2017.
Article in English | MEDLINE | ID: mdl-29224017

ABSTRACT

BACKGROUND/AIMS: Regulatory T cells (Tregs) can suppress immunologic damage in myocardial ischaemia/reperfusion injury (MIRI), however, the isolation and ex vivo expansion of these cells for clinical application remains challenging. Here, we investigated whether the IL-2/anti-IL-2 complex (IL-2C), a mediator of Treg expansion, can attenuate MIRI in mice. METHODS: Myocardial I/R was surgically induced in male C57BL/6 mice, aged 8-10 weeks, that were randomly assigned to 1) sham group (Sham), 2) Phosphate Buffered Saline (PBS), 3) IL-2-anti-IL-2 Ab complex (IL-2C), or 4) sham group, 5) PBS, 6) IL-2C after MIRI, or 7) IL-2C, 8) IL-2C+anti-CD25 mAbs, or 9) IL-2C; 10) IL-2C+anti-TGF-ß1 mAbs, 11) IL-2C+anti-IL-10 mAbs. The following parameters were measured at different time points: infarct area, myocardial apoptosis, splenocytes, the inhibitory function of Tregs, and presence of inflammatory factors. In addition, immunohistochemistry analysis was performed. RESULTS: We observed that Tregs were activated in response to MIRI. IL-2C administered before MIRI induced Treg expansion in both spleen and heart, attenuated Th1 and Th17 cell numbers, improved myocardial function, and attenuated both infiltration of inflammatory cells and apoptosis after MIRI. Furthermore, IL-2C administration reduced expression of inflammatory cytokines in the heart and attenuated proliferation of splenic cells. Depletion of Tregs with anti-CD25 mAb abrogated the beneficial effects of IL-2C. However, IL-2C-mediated myocardial protection was not dependent on either IL-10 or TGF-ß. In addition, IL-2C administration after MIRI did not reduce infarct area, but did improve myocardial function slightly and reduced myocardial fibrosis. CONCLUSION: Our results demonstrate that IL-2C-induced Treg expansion attenuates MIRI and improves myocardial recovery in vivo, suggesting that IL-2C is a promising therapeutic target for myocardial IRI.


Subject(s)
Antigen-Antibody Complex/pharmacology , Interleukin-2/immunology , Myocardial Reperfusion Injury/pathology , T-Lymphocytes, Regulatory/drug effects , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigen-Antibody Complex/therapeutic use , Apoptosis/drug effects , Cytokines/metabolism , Disease Models, Animal , Hemodynamics/drug effects , Interleukin-10/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardium/cytology , Myocardium/immunology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Transforming Growth Factor beta1/immunology
10.
Med Sci Monit ; 23: 2007-2016, 2017 Apr 26.
Article in English | MEDLINE | ID: mdl-28445445

ABSTRACT

BACKGROUND Malvidin (alvidin-3-glucoside) is a polyphenol that belongs to the class of natural anthocyanin, which is abundantly found in red wines, colored fruits, and the skin of red grapes. Therefore, the current investigation was intended to evaluate the effect of malvidin against myocardial infarction induced by isoproterenol in the rats. MATERIAL AND METHODS The cardioprotective effects was assessed by determining the effect of malvidin on the activities of endogenous antioxidants - catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH) - and on the levels of lipid peroxidation and serum marker enzymes. The serum levels of IL-6 and TNF-α were also determined using an enzyme-linked immunosorbent assay (ELISA) kit. RESULTS The present study demonstrated a significant cardioprotective effect of malvidin by restoring the defensive activities of endogenous antioxidants - catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH) - and by reducing the levels of lipid peroxidation and serum marker enzymes lactate dehydrogenase (LD) and creatine kinase (CK). Malvidin significantly ameliorated the histopathological changes and impaired mitochondria in the cardiac necrosis stimulated with isoproterenol. Additionally, the results also demonstrated that nuclear translocation of Nrf-2 and subsequent HO-1 expression might be associated with nuclear factor kappa B (NF-κB) pathway activation. CONCLUSIONS Our findings suggest that malvidin exerts cardioprotective effects that might be due to possible strong antioxidant and anti-inflammatory activities. Therefore, this study provides the basis for the development of malvidin as a safe and effective treatment of myocardial infarction.


Subject(s)
Anthocyanins/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Infarction/prevention & control , Animals , Anthocyanins/pharmacology , Antioxidants/pharmacology , Catalase/drug effects , Catalase/metabolism , Enzyme-Linked Immunosorbent Assay , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Heart/drug effects , Isoproterenol/pharmacology , Lipid Peroxidation/drug effects , Male , Myocardial Infarction/metabolism , Myocardium/pathology , Phytotherapy , Plant Extracts/pharmacology , Rats , Rats, Wistar , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism
11.
Cell Physiol Biochem ; 38(3): 1015-29, 2016.
Article in English | MEDLINE | ID: mdl-26939053

ABSTRACT

BACKGROUND/AIMS: Recent studies have shown that circulating microRNAs (miRNAs) are emerging as promising biomarkers for cardiovascular diseases. This study aimed to determine whether miR-19b-3p, miR-134-5p and miR-186-5p can be used as novel indicators for acute myocardial infarction (AMI). METHODS: To investigate the kinetic expression of the three selected miRNAs, we enrolled 18 patients with AMI and 20 matched controls. Plasma samples were collected from each participant, and total RNA was extracted. Quantitative real-time PCR and ELISA assays were used to investigate the expression of circulating miRNAs and cardiac troponin I (cTnI), respectively. Plasma samples from another age- and gender-matched cohort were collected to investigate the impact of medications for AMI on the expression of the selected miRNAs. RESULTS: Levels of plasma miR-19b-3p, miR-134-5p and miR-186-5p were significantly increased in early stage of AMI. Plasma miR-19b-3p and miR-134-5p levels reached peak expression immediately after admission (T0), whereas miR-186-5p achieved peak expression at 4 h after T0. All of these times were earlier than the peak for cTnI (8 h after T0). In addition, all three miRNAs were positively correlated with cTnI. Receiver Operating Characteristic (ROC) analysis indicated that each single miRNA showed considerable diagnostic efficiency for predicting AMI. Furthermore, combining all three miRNAs in a panel increased the efficiency of distinguishing between patients with AMI and controls. Moreover, we found that heparin and medications for AMI did not impact the expression of these circulating miRNAs. CONCLUSION: Circulating miR-19b-3p, miR-134-5p and miR-186-5p could be considered promising novel diagnostic biomarkers for the early phase of AMI.


Subject(s)
MicroRNAs/blood , Myocardial Infarction/diagnosis , Myocardial Infarction/genetics , Adult , Aged , Early Diagnosis , Female , Gene Expression Regulation , Genetic Markers/genetics , Humans , Male , Middle Aged , Myocardial Infarction/metabolism , ROC Curve , Troponin I/genetics , Troponin I/metabolism
12.
Mol Med ; 21: 143-53, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25685964

ABSTRACT

Valsartan has a protective effect against hypertension and atherosclerosis in humans and experimental animal models. This study aimed to determine the effect of prolonged treatment with angiotensin II (Ang II) on atherosclerosis and the effect of valsartan on the activity of CD4(+) T lymphocyte subsets. The results showed that prolonged treatment (8 wks) with exogenous Ang II resulted in an increased atherosclerotic plaque size and a switch of stable-to-unstable plaque via modulating on CD4(+) T lymphocyte activity, including an increase in the T helper cell type 1 (Th1) and Th17 cells and a decrease in Th2 and regulatory T (Treg) cells. In contrast, valsartan treatment efficiently reversed the imbalance in CD4(+) T lymphocyte activity, ameliorated atherosclerosis and elicited a stable plaque phenotype in addition to controlling blood pressure. In addition, treatment with anti-interleukin (IL)-5 monoclonal antibodies weakened the antiatherosclerotic effects of valsartan without affecting blood pressure.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II/pharmacology , Atherosclerosis/etiology , Th2 Cells/drug effects , Th2 Cells/immunology , Valsartan/pharmacology , Angiotensin II/administration & dosage , Animals , Antibodies, Monoclonal/pharmacology , Apolipoproteins E/deficiency , Atherosclerosis/pathology , Blood Pressure/drug effects , Blood Pressure/genetics , Body Weight , Disease Models, Animal , Inflammation/etiology , Inflammation/pathology , Interleukin-5/antagonists & inhibitors , Lipids/blood , Male , Mice , Mice, Knockout , Plaque, Atherosclerotic/pathology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th2 Cells/metabolism
13.
Cell Physiol Biochem ; 35(1): 213-26, 2015.
Article in English | MEDLINE | ID: mdl-25591764

ABSTRACT

BACKGROUND/AIMS: Hypoxia is a basic pathological challenge that is associated with numerous cardiovascular disorders including aberrant cardiac remodeling. Transforming growth factor beta (TGF-ß) signaling pathway plays a pivotal role in mediating cardiac fibroblast (CF) function and cardiac fibrosis. Recent data suggested that microRNA-101a (miR-101a) exerted anti-fibrotic effects in post-infarct cardiac remodeling and improved cardiac function. This study aimed to investigate the potential relationship between hypoxia, miR-101a and TGF-ß signaling pathway in CFs. METHODS AND RESULTS: Two weeks following coronary artery occlusion in rats, the expression levels of both TGFß1 and TGFßRI were increased, but the expression of miR-101a was decreased at the site of the infarct and along its border. Cultured rat neonatal CFs treated with hypoxia were characterized by the up-regulation of TGFß1 and TGFßRI and the down-regulation of miR-101a. Delivery of miR-101a mimics significantly suppressed the expression of TGFßRI and p-Smad 3, CF differentiation and collagen content of CFs. These anti-fibrotic effects were abrogated by co-transfection with AMO-miR-101a, an antisense inhibitor of miR-101a. The repression of TGFßRI, a target of miR-101a, was validated by luciferase reporter assays targeting the 3'UTR of TGFßRI. Additionally, we found that overexpression of miR-101a reversed the improved migration ability of CFs and further reduced CF proliferation caused by hypoxia. CONCLUSION: Our study illustrates that miR-101a exerts anti-fibrotic effects by targeting TGFßRI, suggesting that miR-101a plays a multi-faceted role in modulating TGF-ß signaling pathway and cardiac fibrosis.


Subject(s)
Cell Hypoxia , Fibroblasts/metabolism , MicroRNAs/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Down-Regulation , Fibroblasts/cytology , Fibrosis/genetics , Male , MicroRNAs/antagonists & inhibitors , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/cytology , Myocardium/pathology , Oligonucleotides, Antisense/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Up-Regulation
14.
Cell Physiol Biochem ; 35(2): 553-70, 2015.
Article in English | MEDLINE | ID: mdl-25612606

ABSTRACT

BACKGROUND: Accumulating evidence shows that the pathological autoreactive immune response is responsible for plaque rupture and the subsequent onset of acute coronary syndrome (ACS). Naturally occurring CD4(+)CD25(+)regulatory T cells (nTregs) are indispensable in suppressing the pathological autoreactive immune response and maintaining immune homeostasis. However, the number and the suppressive function of glycoprotein-A repetitions predominant (GARP) (+) CD4(+) CD25(+) activated nTregs were impaired in patients with ACS. Recent evidence suggests that heme oxygenase-1 (HO-1) can regulate the adaptive immune response by promoting the expression of Foxp3. We therefore hypothesized that HO-1 may enhance the function of GARP(+) CD4(+) CD25(+)Tregs in patients with ACS and thus regulate immune imbalance. METHODS: T lymphocytes were isolated from healthy volunteers (control, n=30) and patients with stable angina (SA, n=40) or ACS (n=51). Half of these cells were treated with an HO-1 inducer (hemin) for 48 h, and the other half were incubated with complete RPMI-1640 medium. The frequencies of T-helper 1 (Th1), Th2, Th17 and latency-associated peptide (LAP) (+)CD4(+) T cells and the expression of Foxp3 and GARP by CD4(+)CD25(+)T cells were then assessed by measuring flow cytometry after stimulation in vitro. The suppressive function of activated Tregs was measured by thymidine uptake. The levels of transforming growth factor-1 (TGF-ß1) in the plasma were measured using enzyme-linked immunosorbent assay (ELISA). The expression levels of the genes encoding these proteins were analyzed by real-time polymerase chain reaction. RESULTS: Patients with ACS exhibited an impaired number and suppressive function of GARP(+) CD4(+) CD25(+)Tregs and a mixed Th1/Th17-dominant T cell response when compared with the SA and control groups. The expression of LAP in T cells was also lower in patients with ACS compared to patients with SA and the control individuals. Treatment with an HO-1 inducer enhanced the biological activity of GARP(+) CD4(+) CD25(+)Tregs and resulted in increased expression of LAP and GARP by activated T cells. CONCLUSIONS: The reduced number and impaired suppressive function of GARP(+) CD4(+) CD25(+)Tregs result in excess effector T cell proliferation, leading to plaque instability and the onset of ACS. HO-1 can effectively restore impaired GARP(+) CD4(+) CD25(+)Tregs from patients with ACS by promoting LAP and GARP expression on activated T cells.


Subject(s)
Acute Coronary Syndrome/immunology , Angina, Stable/immunology , Heme Oxygenase-1/metabolism , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/metabolism , Cells, Cultured , Female , Forkhead Transcription Factors/metabolism , Hemin , Humans , Lymphocyte Activation , Male , Membrane Proteins/metabolism , Middle Aged , T-Lymphocytes, Regulatory/cytology , Up-Regulation
15.
Mediators Inflamm ; 2015: 490120, 2015.
Article in English | MEDLINE | ID: mdl-26819499

ABSTRACT

OBJECTIVE: Recent studies suggest that IL-38 is associated with autoimmune diseases. Furthermore, IL-38 is expressed in human atheromatous plaque. However, the plasma levels of IL-38 in patients with ST-segment elevation myocardial infarction (STEMI) have not yet to be investigated. METHODS: On admission, at 24 h, at 48 h, and at 7 days, plasma IL-38, C-reactive protein (CRP), cardiac troponin I (cTNI), and N-terminal of the prohormone brain natriuretic peptide (NT-proBNP) levels were measured and IL-38 gene in peripheral blood mononuclear cells (PBMCs) was detected in STEMI patients. RESULTS: The results showed that plasma IL-38 levels and IL-38 gene expression in PBMCs were significantly increased in STEMI patients compared with control group and were time dependent, peaked at 24 h. In addition, plasma IL-38 levels were dramatically reduced in patients with reperfusion treatment compared with control group. Similar results were also demonstrated with CRP, cTNI, and NT-proBNP levels. Furthermore, IL-38 levels were found to be positively correlated with CRP, cTNI, and NT-proBNP and be weakly negatively correlated with left ventricular ejection fraction (LVEF) in STEMI patients. CONCLUSIONS: The results indicate that circulating IL-38 is a potentially novel biomarker for patients with STEMI and IL-38 might be a new target for MI study.


Subject(s)
Interleukins/blood , Myocardial Infarction/immunology , Adult , Aged , Biomarkers , C-Reactive Protein/analysis , Electrocardiography , Female , Humans , Male , Middle Aged , Myocardial Infarction/therapy , Myocardial Reperfusion , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Troponin I/blood
16.
Mediators Inflamm ; 2015: 479123, 2015.
Article in English | MEDLINE | ID: mdl-26688617

ABSTRACT

AIMS: miR-208a is associated with adverse outcomes in several cardiac pathologies known to have increased apoptosis, including myocardial infarction (MI). We investigated if miR-208a has proapoptotic effects on ischemic cardiomyocytes and if its silencing has therapeutic benefits in MI. METHODS AND RESULTS: The effect of miR-208a on apoptosis during ischemia was studied in cultured neonatal mice myocytes transfected with agomir or antagomir. Differential gene expression was assessed using microarrays. MI was induced in male C57BL/6 mice randomly assigned to antagomir (n = 6) or control group (n = 7), while sham group (n = 7) had sham operation done. Antagomir group received miR208a antagomir, while control and sham group mice received vehicle only. At 7 and 28 days, echocardiography was done and thereafter hearts were harvested for analysis of apoptosis by TUNEL method, fibrosis using Masson's trichrome, and hypertrophy using hematoxylin and eosin. miR-208a altered apoptosis genes expression and increased apoptosis in ischemic cardiomyocytes. Therapeutic inhibition of miR-208a decreased cardiac fibrosis, hypertrophy, and apoptosis and significantly improved cardiac function 28 days after MI. CONCLUSION: miR-208a alters apoptosis genes expression and promotes apoptosis in ischemic cardiomyocytes, and its silencing attenuates apoptosis, fibrosis, and hypertrophy after MI, with significant improvement in cardiac function.


Subject(s)
Apoptosis , Gene Expression Regulation , MicroRNAs/physiology , Myocardial Infarction/therapy , Myocardial Ischemia/pathology , Myocytes, Cardiac/pathology , Animals , Cells, Cultured , Fibrosis , Gene Silencing , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology
17.
Mediators Inflamm ; 2015: 457167, 2015.
Article in English | MEDLINE | ID: mdl-26663989

ABSTRACT

BACKGROUND: Recently, several studies suggest that galectin-9 (Gal-9) might play a pivotal role in the pathogenesis of autoimmune diseases. However, the exact role of Gal-9 in atherosclerosis remains to be elucidated. METHODS: Serum Gal-9, high-sensitivity C-reactive protein (hs-CRP), interferon- (IFN-) γ, interleukin- (IL-) 4, IL-17, and transforming growth factor- (TGF-) ß1 were measured. The effect of Gal-9 on peripheral blood mononuclear cells (PBMC) was investigated in patients with normal coronary artery (NCA). RESULTS: The lowest level of Gal-9 was found in the ST-segment elevation myocardial infarction (STEMI) group, followed by the non-ST-segment elevation ACS (NSTEACS), the NCA, and the stable angina pectoris (SAP) groups, respectively. Additionally, Gal-9 was found to be independently associated with hs-CRP, lipoprotein(a), and creatinine. Notably, Gal-9 was also noted to be an independent predictor of the Gensini score. Moreover, Gal-9 suppressed T-helper 17 (Th17) and expanded regulatory T cells (Tregs), resulting in decreased IL-17 production and increased secretion of TGF-ß1. CONCLUSIONS: Serum Gal-9 is associated with not only coronary artery disease (CAD), but also the severity of coronary arteries stenosis. Gal-9 can expand Tregs and suppress Th17 development in activated PBMC, implying that Gal-9 has the potential to dampen the development of atherosclerosis and may be a new therapy for CAD.


Subject(s)
Coronary Artery Disease/etiology , Galectins/blood , Adult , Aged , C-Reactive Protein/analysis , Coronary Artery Disease/blood , Cytokines/blood , Female , Humans , Male , Middle Aged , Regression Analysis
18.
Mediators Inflamm ; 2015: 563951, 2015.
Article in English | MEDLINE | ID: mdl-26161004

ABSTRACT

Chemerin15 (C15), an endogenous anti-inflammatory component, inhibits the activity of neutrophils and macrophages through G protein-coupled receptor ChemR23; however, its role as well as functional mechanism in mouse myocardial ischemia/reperfusion (I/R) injury remains unknown. Methods. Sham or I/R operations were performed on C57BL/6J mice. The I/R mice received an injection of C15 immediately before reperfusion. Serum troponin T levels, infarct size, cardiomyocyte apoptosis, reactive oxygen species (ROS) production, and infiltration of neutrophils were assessed 24 h after reperfusion, while the macrophage phenotypes, macrophage infiltration, and inflammatory cytokine levels were assessed 48 h after reperfusion. Results. Compared with the control group, the C15-treated mice showed an obvious amelioration of I/R injury and displayed less ROS, accompanied by reduced neutrophil recruitment. C15 decreased the tumor necrosis factor- (TNF-) α and interleukin- (IL-) 6 levels and increased the IL-10 levels in the serum of the I/R mice, which suggested a suppressed inflammatory response that could be related to elevated alternatively activated M2 macrophages with characteristic skewed expression of M2 markers and inhibition of classically activated M1 marker expression. Conclusion. C15 may induce alternatively activated M2 macrophage polarization and suppress the inflammatory response to protect against myocardial I/R injury in mice.


Subject(s)
Chemotactic Factors/therapeutic use , Intercellular Signaling Peptides and Proteins/therapeutic use , Macrophage Activation/drug effects , Myocardial Reperfusion Injury/prevention & control , Peptide Fragments/therapeutic use , Animals , Apoptosis/drug effects , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Neutrophil Infiltration/drug effects
19.
Mediators Inflamm ; 2015: 841472, 2015.
Article in English | MEDLINE | ID: mdl-26063978

ABSTRACT

Regulatory T cells play an important role in the progression of atherosclerosis. GARP is a newly biological membrane molecule existed on activated Tregs, which is related to the release of TGF-ß. The antiatherosclerosis effects of statins partly depend on their multiple immune modulatory potencies. In this paper, we present that atorvastatin could upregulate the expression of GARP and TGF-ß in CD4+ T cells and increase the numbers of CD4+LAP+ and CD4+Foxp3+ regulatory T cells in ApoE-/- mice. Also, we indicate that atorvastatin promotes the aggregation of GARP+ and Foxp3+ cells and secretory of the TGF-ß1 in atherosclerotic plaques. Furthermore, we prove that atorvastatin could delay the procession of atherosclerosis and improve the stability of atherosclerotic plaques. Interestingly, we report that inhibition of GARP distinctly inhibits the anti-inflammatory effects of atorvastatin. We conclude that atorvastatin improves the inflammatory response in atherosclerosis partly by upregulating the expression of GARP on regulatory T cells.


Subject(s)
Atherosclerosis/drug therapy , Atherosclerosis/immunology , Atorvastatin/therapeutic use , Membrane Proteins/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis/drug effects , Atherosclerosis/metabolism , Blotting, Western , Cells, Cultured , Flow Cytometry , Immunohistochemistry , Male , Membrane Proteins/genetics , Mice , T-Lymphocytes, Regulatory
20.
Cell Physiol Biochem ; 33(3): 621-32, 2014.
Article in English | MEDLINE | ID: mdl-24603196

ABSTRACT

BACKGROUND: Atherosclerosis (AS) is an inflammatory and immune disease. Regulatory T cells (Tregs) suppress the activation of T cells and have been shown to play a protective role during the pathogenesis of AS. However, specific markers for Tregs are lacking. Recently, glycoprotein A repetitions predominant (GARP) was discovered as a specific marker of activated Tregs, and we therefore utilized GARP as a specific surface marker for Tregs in the current study. METHODS: To assess whether GARP(+) Tregs are downregulated in patients with acute coronary syndrome (ACS), we examined CD4(+)CD25(+)GARP(+) T cell frequencies as well as their associated cytokines and suppressive function. Additionally, we compared GARP expression to that of FOXP3, which may be more sensitive as a marker of activated Tregs in patients with ACS. RESULTS: Patients with ACS demonstrated a significant decrease in circulating CD4(+)CD25(+)GARP(+) Tregs. Moreover, the suppressive function of Tregs and levels of related cytokines were also impaired in ACS patients compared to those with stable angina (SA) or normal coronary artery (NCA). Additionally, after TCR stimulation, peripheral blood mononuclear cells (PBMCs) from patients with ACS exhibited a decrease in CD4(+)CD25(+)GARP(+) Tregs. CONCLUSIONS: These fnding indicate that circulating CD4(+)CD25(+)GARP(+) Tregs are impaired in patients withACS. Thus, targeting GARP may promote the protective function of Tregs in ACS.


Subject(s)
Acute Coronary Syndrome/immunology , Membrane Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/pathology , Aged , Female , Forkhead Transcription Factors/blood , Forkhead Transcription Factors/immunology , Gene Expression Regulation/immunology , Humans , Interleukin-2 Receptor alpha Subunit/blood , Interleukin-2 Receptor alpha Subunit/immunology , Male , Membrane Proteins/blood , Middle Aged , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
SELECTION OF CITATIONS
SEARCH DETAIL