Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Acta Pharmacol Sin ; 42(12): 2046-2057, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33623121

ABSTRACT

Nuclear factor kappa B (NF-κB) activation contributes to many vascular inflammatory diseases. The present study tested the hypothesis that microRNA-17-3p (miR-17-3p) suppresses the pro-inflammatory responses via NF-κB signaling in vascular endothelium. Human umbilical vein endothelial cells (HUVECs), transfected with or without miR-17-3p agomir/antagomir, were exposed to lipopolysaccharide (LPS), and the inflammatory responses were determined. The cellular target of miR-17-3p was examined with dual-luciferase reporter assay. Mice were treated with miR-17-3p agomir and the degree of LPS-induced inflammation was determined. In HUVECs, LPS caused upregulation of miR-17-3p. Overexpression of miR-17-3p in HUVECs inhibited NIK and IKKß binding protein (NIBP) protein expression and suppressed LPS-induced phosphorylation of inhibitor of kappa Bα (IκBα) and NF-κB-p65. The reduced NF-κB activity was paralleled by decreased protein levels of NF-κB-target gene products including pro-inflammatory cytokine [interleukin 6], chemokines [interleukin 8 and monocyte chemoattractant protein-1] and adhesion molecules [vascular cell adhesion molecule-1, intercellular adhesion molecule-1 and E-selectin]. Immunostaining revealed that overexpression of miR-17-3p reduced monocyte adhesion to LPS-stimulated endothelial cells. Inhibition of miR-17-3p with antagomir has the opposite effect on LPS-induced inflammatory responses in HUVECs. The anti-inflammatory effect of miR-17-3p was mimicked by NIBP knockdown. In mice treated with LPS, miR-17-3p expression was significantly increased. Systemic administration of miR-17-3p for 3 days suppressed LPS-induced NF-κB activation and monocyte adhesion to endothelium in lung tissues of the mice. In conclusion, miR-17-3p inhibits LPS-induced NF-κB activation in HUVECs by targeting NIBP. The findings therefore suggest that miR-17-3p is a potential therapeutic target/agent in the management of vascular inflammatory diseases.


Subject(s)
Endothelium, Vascular/metabolism , I-kappa B Kinase/metabolism , Inflammation/metabolism , MicroRNAs/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factor RelA/metabolism , Animals , Antagomirs/pharmacology , Cell Adhesion Molecules/metabolism , Cytokines/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/chemically induced , Lipopolysaccharides , Male , Mice , NF-KappaB Inhibitor alpha/metabolism , Signal Transduction/physiology , Up-Regulation/physiology , NF-kappaB-Inducing Kinase
2.
J Leukoc Biol ; 97(4): 797-805, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25722319

ABSTRACT

There has been increasing evidence that chronic immune activation plays critical roles in the pathogenesis of DCM. CD4(+) LAP(+) Tregs are a newly identified T cell subset with suppressive function on the immune response. This study was designed to investigate whether the circulating frequency and function of CD4(+)LAP(+) Tregs would be impaired in patients with DCM. The results demonstrated that DCM patients had a significantly lower frequency of circulating CD4(+)LAP(+) Tregs compared with control donors. CD4(+)LAP(+) Tregs from DCM patients showed compromised function to suppress proliferation of CD4(+) LAP(-)CD25(int/low) T cells and proliferation and IgG production of B cells. Moreover, B cell proliferation and IgG subset production could be directly suppressed by CD4(+) LAP(+) Tregs. TGF-ß and contact-dependent mechanisms were involved in CD4(+)LAP(+) Treg-mediated suppression. Correlation analysis suggested that CD4(+)LAP(+) Treg frequency was positively correlated with LVEF and negatively correlated with serum IgG3 and NT-proBNP concentration in patients with DCM. Our results are the first to demonstrate that the frequencies of CD4(+)LAP(+) Tregs in patients with DCM are reduced and that their suppressive function is compromised. Defective CD4(+) LAP(+) Tregs may be an underlying mechanism of immune activation in DCM patients.


Subject(s)
Cardiomyopathy, Dilated/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta1/blood , Adult , Aged , B-Lymphocytes/immunology , CD4 Lymphocyte Count , Cardiomyopathy, Dilated/blood , Cardiomyopathy, Dilated/diagnostic imaging , Cardiomyopathy, Dilated/physiopathology , Flow Cytometry , Humans , Immune Tolerance , Immunoglobulin G/blood , Lymphocyte Activation/immunology , Lymphocyte Cooperation/immunology , Middle Aged , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Stroke Volume , T-Lymphocyte Subsets/chemistry , T-Lymphocytes, Regulatory/chemistry , Transforming Growth Factor beta/pharmacology , Ultrasonography
3.
Int J Clin Exp Pathol ; 8(11): 14050-62, 2015.
Article in English | MEDLINE | ID: mdl-26823719

ABSTRACT

MicroRNA (miRNA, miR)-155 is the most promising pro-inflammatory miRNA molecule. Lipopolysaccharide (LPS) and oxidized low-density lipoprotein (oxLDL) are the most well-known foreign antigens, initiating immune responses against infection and the development of atherosclerosis (AS), respectively. To explore whether miR-155 is involved in regulating LPS- and oxLDL-initiated inflammations, we investigated the level of miR-155 in both LPS- and oxLDL-treated RAW264.7 cells, assessed whether miR-155 induce morphologic changes of the cells and how did it regulate the production of surface markers and cytokines. The results showed that the level of miR-155 was significantly increased by LPS and was modestly increased by oxLDL. Moreover, RAW264.7 cells displayed morphological transformations from macrophage-like cells into DC-like cells when miR-155 was over-expressed. Furthermore, the gain- and loss-of-function studies demonstrated that miR-155 induced the expression of the surface markers (including MHC-II, MHC-I, CD86, and CD83) and pro-inflammatory cytokines (including interleukin (IL)-12, IL-6, and IL-1b) in both LPS- and oxLDL-treated RAW264.7 cells. Additionally, miR-155 induced the expression of CD36 in oxLDL-treated RAW264.7 cells. In conclusion, up-regulated miR-155 is able to induce morphological and phenotypic changes, and the expression of pro-inflammatory cytokines in both LPS- and oxLDL-treated RAW264.7 cells. Therefore, our study suggests that miR-155 is one important regulator involved in enhancing both LPS- and oxLDL-initiated inflammations, which is critical for the progression of immune responses as well as for the development of AS.


Subject(s)
Cell Transdifferentiation , Dendritic Cells/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Animals , CD36 Antigens/metabolism , Cell Shape , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Lipoproteins, LDL/pharmacology , Macrophages/drug effects , Macrophages/immunology , Mice , MicroRNAs/genetics , Phenotype , RAW 264.7 Cells , Time Factors , Up-Regulation
4.
J Mol Med (Berl) ; 92(10): 1105-16, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24965614

ABSTRACT

Inflammatory responses play an important role in the pathogenesis of adverse ventricular remodeling after myocardial infarction (MI). We previously demonstrated that interleukin (IL)-17A plays a pathogenic role in myocardial ischemia/reperfusion injury and viral myocarditis. However, the role of IL-17A in post-MI remodeling and the related mechanisms have not been fully elucidated. Acute MI was induced by permanent ligation of the left anterior descending coronary artery in C57BL/6 mice. Repletion of IL-17A significantly aggravated both early- and late-phase ventricular remodeling, as demonstrated by increased infarct size, deteriorated cardiac function, increased myocardial fibrosis, and cardiomyocyte apoptosis. By contrast, genetic IL-17A deficiency had the opposite effect. Additional studies in vitro indicated that IL-17A induces neonatal cardiomyocyte (from C57BL/6 mice) apoptosis through the activation of p38, p53 phosphorylation, and Bax redistribution. These data demonstrate that IL-17A induces cardiomyocyte apoptosis through the p38 mitogen-activated protein kinase (MAPK)-p53-Bax signaling pathway and promotes both early- and late-phase post-MI ventricular remodeling. IL-17A might be an important target in preventing heart failure after MI. Key message: We demonstrated that IL-17A plays a pathogenic role both in the early and late stages of post-MI remodeling. IL-17A induces murine cardiomyocyte apoptosis. IL-17A induces murine cardiomyocyte apoptosis through the p38 MAPK-p53-Bax signaling pathway.


Subject(s)
Interleukin-17/metabolism , Myocardial Infarction/metabolism , Ventricular Remodeling/physiology , Animals , Apoptosis/drug effects , Cytochromes c/metabolism , Fibrosis , Interleukin-17/genetics , Male , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Messenger/metabolism , Receptors, Interleukin-17/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Cell Physiol Biochem ; 32(5): 1265-74, 2013.
Article in English | MEDLINE | ID: mdl-24247374

ABSTRACT

BACKGROUND: Interleukin (IL)-17A, a newly identified cytokine, may participate in the transition of a stable plaque into an unstable plaque. Macrophages play a critical role in the destabilization of atherosclerotic plaque. METHODS: RAW 264.7 cells were stimulated with IL-17A. The mRNA expression of inflammatory cytokines was determined by RT-PCR. The cytokines production in the supernatants was measured by ELISA. Small interfering RNA (siRNA) was used to confirm that IL-17A-induced pro-inflammatory cytokines production via IL-17RA signaling. The western blot assay was used to detect the phosphorylation of MAPKinases including p38 and ERK1/2. The DNA binding activity of nuclear factor NF-κB and AP-1 were detected by EMSA. RESULTS: IL-17A induced the production of pro-inflammatory cytokines in macrophages in a time- and dose-dependent manner, such as tumor necrosis factor (TNF)-α, IL-1ß, and IL-6. Meanwhile, IL-17A resulted in the phosphorylation of p38 and ERK1/2 and increased DNA-binding activity of NF-κB and AP-1. Pharmacological inhibitors of p38 and ERK1/2 partly attenuated IL-17A-induced TNF-α, IL-1ß, and IL-6 production. Either NF-κB inhibitor or AP-1 inhibitor also partly decreased the IL-17A-induced cytokine production. CONCLUSIONS: IL-17A induces pro-inflammatory cytokines production in macrophages via MAPKinases, NF-κB and AP-1 pathway.


Subject(s)
Cytokines/metabolism , Inflammation/metabolism , Interleukin-17/pharmacology , Macrophages/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Animals , Cell Line , Cytokines/genetics , Enzyme Inhibitors/pharmacology , Interleukin-17/metabolism , Interleukin-1beta/genetics , Interleukin-6/genetics , Macrophages/drug effects , Mice , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, Interleukin-17/metabolism , Tumor Necrosis Factor-alpha/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
6.
J Biol Chem ; 287(41): 34157-66, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22872639

ABSTRACT

Regulatory T (Treg) cells play a protective role against the development of atherosclerosis. Previous studies have revealed Treg cell defects in patients with non-ST elevation acute coronary syndrome (NSTACS), but the mechanisms underlying these defects remain unclear. In this study, we found that the numbers of peripheral blood CD4(+)CD25(+)CD127(low) Treg cells and CD4(+)CD25(+)CD127(low)CD45RA(+)CD45RO(-) naive Treg cells were lower in the NSTACS patients than in the chronic stable angina (CSA) and the chest pain syndrome (CPS) patients. However, the number of CD4(+)CD25(+)CD127(low)CD45RA(-)CD45RO(+) memory Treg cells was comparable in all of the groups. The frequency of CD4(+)CD25(+)CD127(low)CD45RO(-)CD45RA(+)CD31(+) recent thymic emigrant Treg cells and the T cell receptor excision circle content of purified Treg cells were lower in the NSTACS patients than in the CSA patients and the CPS controls. The spontaneous apoptosis of Treg cells (defined as CD4(+)CD25(+)CD127(low)annexin V(+)7-AAD(-)) was increased in the NSTACS patients compared with the CSA and CPS groups. Furthermore, oxidized LDL could induce Treg cell apoptosis, and the oxidized LDL levels were significantly higher in the NSTACS patients than in the CSA and CPS groups. In accordance with the altered Treg cell levels, the concentration of TNF-α was increased in the NSTACS patients, resulting in a decreased IL-10/TNF-α ratio. These findings indicate that the impaired thymic output of Treg cells and their enhanced susceptibility to apoptosis in the periphery were responsible for Treg cell defects observed in the NSTACS patients.


Subject(s)
Acute Coronary Syndrome/blood , Apoptosis , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/metabolism , Acute Coronary Syndrome/genetics , Acute Coronary Syndrome/immunology , Aged , Antigens, CD/blood , Antigens, CD/immunology , Biological Transport/immunology , Female , Humans , Interleukin-11/blood , Interleukin-11/immunology , Lipoproteins, LDL/blood , Lipoproteins, LDL/immunology , Male , Middle Aged , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Thymus Gland/immunology , Thymus Gland/pathology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
7.
Basic Res Cardiol ; 107(1): 232, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22189560

ABSTRACT

Persistent inflammatory responses participate in the pathogenesis of adverse ventricular remodeling after myocardial infarction (MI). We hypothesized that regulatory T (Treg) cells modulate inflammatory responses, attenuate ventricular remodeling and subsequently improve cardiac function after MI. Acute MI was induced by ligation of the left anterior descending coronary artery in rats. Infiltration of Foxp3(+) Treg cells was detected in the infarcted heart. Expansion of Treg cells in vivo by means of adoptive transfer as well as a CD28 superagonistic antibody (JJ316) resulted in an increased number of Foxp3(+) Treg cells in the infarcted heart. Subsequently, rats with MI showed improved cardiac function following Treg cells transfer or JJ316 injection. Interstitial fibrosis, myocardial matrix metalloproteinase-2 activity and cardiac apoptosis were attenuated in the rats that received Treg cells transfer. Infiltration of neutrophils, macrophages and lymphocytes as well as expression of tumor necrosis factor (TNF)-α and interleukin (IL)-1ß were also significantly decreased, and the CD8(+) cardiac-specific cytotoxic T lymphocyte response was inhibited. Expression of interleukin (IL)-10 in the heart, however, was increased. Additional studies in vitro indicated that Treg cells directly protect neonatal rat cardiomyocytes against LPS-induced apoptosis, and this protection depends on the cell-cell contact and IL-10 expression. Furthermore, Treg cells inhibited proinflammatory cytokines production by cardiomyocytes. These data demonstrate that Treg cells serve to protect against adverse ventricular remodeling and contribute to improve cardiac function after myocardial infarction via inhibition of inflammation and direct protection of cardiomyocytes.


Subject(s)
Myocardial Infarction/immunology , T-Lymphocytes, Regulatory/physiology , Ventricular Remodeling/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal , CD8-Positive T-Lymphocytes/physiology , Interleukin-10/physiology , Male , Myocytes, Cardiac/immunology , Rats , Rats, Inbred Lew
8.
PLoS One ; 6(9): e24272, 2011.
Article in English | MEDLINE | ID: mdl-21935395

ABSTRACT

OBJECTIVE: Animal studies suggest that regulatory T (T(reg)) cells play a beneficial role in ventricular remodeling and our previous data have demonstrated defects of T(reg) cells in patients with chronic heart failure (CHF). However, the mechanisms behind T(reg-)cell defects remained unknown. We here sought to elucidate the mechanism of T(reg-)cell defects in CHF patients. METHODS AND RESULTS: We performed flow cytometry analysis and demonstrated reduced numbers of peripheral blood CD4(+)CD25(+)FOXP3(+)CD45RO(-)CD45RA(+) naïve T(reg) (nT(reg)) cells and CD4(+)CD25(+)FOXP3(+)CD45RO(+)CD45RA(-) memory T(reg) (mT(reg)) cells in CHF patients as compared with non-CHF controls. Moreover, the nT(reg)/mT(reg) ratio (p<0.01), CD4(+)CD25(+)FOXP3(+)CD45RO(-) CD45RA(+)CD31(+) recent thymic emigrant T(reg) cell (RTE-T(reg)) frequency (p<0.01), and T-cell receptor excision circle levels in T(reg) cells (p<0.01) were lower in CHF patients than in non-CHF controls. Combined annexin-V and 7-AAD staining showed that peripheral T(reg) cells from CHF patients exhibited increased spontaneous apoptosis and were more prone to interleukin (IL)-2 deprivation- and CD95 ligand-mediated apoptosis than those from non-CHF individuals. Furthermore, analyses by both flow cytometry and real-time polymerase chain reaction showed that T(reg)-cell frequency in the mediastinal lymph nodes or Foxp3 expression in hearts of CHF patients was no higher than that of the non-CHF controls. CONCLUSION: Our data suggested that the T(reg)-cell defects of CHF patients were likely caused by decreased thymic output of nascent T(reg) cells and increased susceptibility to apoptosis in the periphery.


Subject(s)
Heart Failure/immunology , Heart Failure/physiopathology , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/metabolism , Adult , Apoptosis/genetics , Apoptosis/physiology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chronic Disease , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Heart Failure/metabolism , Humans , Interleukin-2/genetics , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Middle Aged , Real-Time Polymerase Chain Reaction
9.
J Lipid Res ; 52(5): 1023-32, 2011 May.
Article in English | MEDLINE | ID: mdl-21368320

ABSTRACT

In this study, we investigated the hypothesis that regulatory T cells (T(reg)) are involved in the immunomodulatory effects of statins on rheumatoid arthritis (RA) patients. The 12-week study cohort consisted of 55 RA patients and 42 control subjects allocated to either a group treated with atorvastatin (AT) (20 mg/day) or a non-AT group. T(reg) numbers, suppressive function, serum inflammatory markers, and disease activity were evaluated before and after the therapy. Furthermore, the effects of AT on the frequency and suppressive function of T(reg) were determined in vitro. Our data revealed that the suppressive function of T(reg) from RA patients significantly decreased compared with that of control subjects. AT significantly reduced erythrosedimentation, C-reactive protein, and disease activity. Concomitantly, T(reg) numbers and suppressive functions were significantly improved by AT. Consistent with the in vivo experiments, AT promoted the generation of T(reg) from primary T cells and enhanced preexisting T(reg) function in vitro. Moreover, we showed that PI3K-Akt-mTOR and ERK signal pathways were involved in the induction of T(reg) by AT. In conclusion, AT significantly increased T(reg) numbers and restored their suppressive function in the RA patients, and this may be relevant in the modulation of uncontrolled inflammation in this disorder.


Subject(s)
Anticholesteremic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Heptanoic Acids/therapeutic use , Pyrroles/therapeutic use , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Adult , Atorvastatin , Female , Humans , Male , Middle Aged , Signal Transduction/drug effects
10.
Atherosclerosis ; 215(2): 471-4, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21300351

ABSTRACT

OBJECTIVE: To determine the effects of interleukin (IL)-17A inhibition on experimental atherosclerosis. METHODS AND RESULTS: ApoE(-/-) mice were treated with either rat anti-mouse IL-17A, mouse anti-mouse IL-17A or isotype-matched control antibodies for 12 weeks (n=8-10 per group). Ldlr(-/-) mice were transplanted with IL-17A-deficient or wild type bone marrow (n=8 per group). Rat anti-mouse IL-17A treatment obviously reduced plaque size by 43% (p<0.01) without evidence of reduced IL-17A signaling. In contrast, mouse anti-mouse IL-17A treatment and IL-17A deficiency in bone marrow cells did not alter lesion size despite significant reduction of IL-17A production. CONCLUSIONS: Inhibition of IL-17A signaling does not alter lesion development in Th1-biased C57BL/6 ApoE(-/-) and Ldlr(-/-) mice with already low levels of IL-17A production. Alteration of lesion development after repeated injections of rat anti-mouse IL-17A antibody in ApoE(-/-) mice could not be attributed to blockade of IL-17A signaling.


Subject(s)
Atherosclerosis/immunology , Interleukin-17/antagonists & inhibitors , Animals , Antibodies/pharmacology , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Female , Interleukin-17/immunology , Interleukin-17/physiology , Male , Mice , Mice, Inbred C57BL , Plaque, Atherosclerotic/prevention & control , Rats , Receptors, LDL/deficiency , Signal Transduction/drug effects
11.
Cell Physiol Biochem ; 25(4-5): 451-8, 2010.
Article in English | MEDLINE | ID: mdl-20332626

ABSTRACT

AIMS: Increasing evidences confirm the role of immune activation in the pathogenesis of chronic heart failure (CHF). Regulatory T cells appear central to the control of immune homeostasis. We assessed the hypothesis that the circulating frequency and function of CD4+CD25+ Foxp3+CD127(low) T regulatory cells (Tregs) would be deranged in patients with CHF. METHODS: Ninety-nine CHF patients due to non-ischemic (NIHF) or ischemic etiology (IHF) and 24 control donors were enrolled in the study. Frequency of circulating Tregs was evaluated by flow cytometry. Foxp3 in peripheral blood mononuclear cells (PBMCs) was assayed at the mRNA level by real-time PCR. Functional properties of Tregs to suppress proliferation and pro-inflammatory cytokines secretion of activated CD4+CD25(-) T cells were measured by proliferation assay and ELISA. RESULTS: The results demonstrated that CHF patients had significantly lower frequency of circulating Tregs and reduced Foxp3 expression in PBMCs compared with control donors. Moreover, Tregs from CHF patients showed compromised function to suppress CD4+CD25(-) T cells proliferation and pro-inflammatory cytokines secretion. A similar pattern with reduced Tregs frequency and compromised function was found in both NIHF and IHF patients. Correlation analysis suggested that Tregs frequency and function positively correlated with LVEF, whereas negatively correlated with LVEDD and NT-proBNP in patients with CHF. CONCLUSIONS: Our data are the first to demonstrate that frequencies of circulating Tregs in patients with CHF are reduced and their suppressive function compromised independently of the etiology. Defective Tregs may be an underlying mechanism of immune activation in CHF patients.


Subject(s)
Forkhead Transcription Factors/metabolism , Heart Failure/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , T-Lymphocytes, Regulatory/immunology , Adult , Chronic Disease , Female , Flow Cytometry , Humans , Male , Middle Aged
12.
Cytokine ; 49(2): 185-93, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19836260

ABSTRACT

OBJECTIVE: Atherosclerosis is a chronic inflammatory disease regulated by T lymphocyte subsets. Recently, CD4(+) CD25(+) Foxp3(+) regulatory T (Treg) cells and Th17 cells have been described as two distinct subsets and have the opposite effects on autoimmunity. Clinical observation has revealed that the Th17/Treg imbalance exists in patients with acute coronary syndrome. We investigated whether the Th17/Treg functional imbalance existed during atherogenesis in ApoE(-/-) mice. METHODS AND RESULTS: Th17/Treg functions at different levels including cell frequencies, related cytokine secretion and key transcription factors were investigated comparatively between ApoE(-/-) mice and their age-matched C57BL/6J mice. The results demonstrated that ApoE(-/-) mice revealed significantly increased secretion of Th17 related cytokines (IL-17 and IL-6) and expression of transcription factor (RORgammat) levels and obviously decreased number in Treg cells, secretion of Treg related cytokines (TGF-beta(1)) and expression of transcription factor (Foxp3) levels as compared with age-matched C57BL/6J mice. Th17 related mediators reached their maximum expression values at the early stage (8-16weeks of age) in ApoE(-/-) mice, and then followed by continuous depression of their expression levels. Meanwhile, the expression of Treg related mediators was much lower in ApoE(-/-) mice than in their age-matched wild-type littermates. CONCLUSIONS: Th17/Treg functional imbalance exists during atherogenesis in ApoE(-/-) mice, suggesting a potential role of Th17/Treg imbalance in the formation and progression of atherosclerosis.


Subject(s)
Apolipoproteins E/immunology , Atherosclerosis/metabolism , Interleukin-17/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Cytokines/genetics , Cytokines/immunology , Humans , Interleukin-17/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Regulatory/cytology , Th1 Cells/cytology
13.
Int J Cardiol ; 139(2): 113-22, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-19028402

ABSTRACT

OBJECTIVE: Atherosclerosis is a chronic inflammatory disease in which the renin-angiotensin-aldosterone system plays an important role. Evidence indicate that the angiotensin type 1 receptor blockers can suppress atherogenesis, but the exact mechanisms have not been fully elucidated. The study was undertaken to investigate the potential effects and molecular mechanisms of an angiotensin type 1 receptor blocker irbesartan on atherogenesis in high cholesterol-diet apolipoprotein E knock-out mice. METHODS AND RESULTS: Adult male apolipoprotein E knock-out mice were given normal diet or high cholesterol-diet and randomized to receive no treatment or irbesartan 10 mg kg(-1) d(-1) for 12 weeks. The apolipoprotein E knock-out mice with high cholesterol-diet were associated with a marked increase in atherosclerotic lesion area, plasma lipid and angiotensin II levels, as well as the expressions of angiotensin type 1 receptor in the aorta. High cholesterol-diet feeding increases the activity of NADPH oxidase subunits (p47(phox) and Rac), extracellular signal-regulated kinase 1/2, janus kinase 2, signal transducer and activator of transcription 3, nuclear factor-kappaB and the expression of tumor necrosis factor-alpha, interleukin 6, monocyte chemoattactant protein-1 and vascular cell adhesion molecule-1 in the aortas. These changes were suppressed in mice that were treated with irbesartan 10 mg kg(-1) d(-1), with no significant change in systolic blood pressure and plasma lipid levels. CONCLUSIONS: The results suggest that irbesartan can attenuate atherosclerosis, and this effect is partly related to the inhibition of oxidative stress and inflammatory signal transduction pathways which eventually leads to the decrease in the expression of inflammatory cytokines.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Biphenyl Compounds/pharmacology , Tetrazoles/pharmacology , Animals , Aorta/metabolism , Aorta/physiology , Atherosclerosis/immunology , Blood Pressure/drug effects , Blood Pressure/physiology , Cholesterol, Dietary/pharmacology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Hypercholesterolemia/drug therapy , Hypercholesterolemia/immunology , Hypercholesterolemia/metabolism , Irbesartan , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Oxidative Stress/physiology , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Vasculitis/drug therapy , Vasculitis/immunology , Vasculitis/metabolism
14.
J Atheroscler Thromb ; 16(5): 641-53, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19776495

ABSTRACT

AIM: Hyperhomocysteinemia (Hhcy) is an important and independent risk factor for atherosclerosis. Recent studies have shown that Poly (ADP-ribose) polymerase (PARP) activation may be associated with Hhcy-induced endothelial dysfunction, which is an important mechanism for Hhcy to affect atherosclerotic progress. Thus, we investigated whether PARP inhibitors may attenuate atheroscle-rotic plaque development in an Hhcy-induced experimental animal model with atherosclerosis. METHODS: Six-week-old homozygous apolipoprotein E-deficient (ApoE-/-) male mice fed a normal diet or high methionine diet randomly received intraperitoneal injections of 10 mg/kg 3-aminoben-zamide (3-AB, a PARP inhibitor) dissolved in phosphate-buffered saline (PBS), or physiological saline every other day for 12 weeks. Atherosclerotic lesion sizes and PARP activity were measured. Related inflammatory factors in atherogenesis were investigated by real-time quantitative PCR and Western blot analysis. RESULTS: Our data demonstrated that ApoE-/- mice fed a high methionine diet generated Hhcy, which subsequently increased the atherosclerotic lesion size significantly, promoted oxidative stress-associated DNA damage and PARP activation, then increased the expression of proinflammatory fac-tors within atherosclerotic plaques. Although PARP inhibition by 3-AB did not markedly inhibit plaque development in ApoE-/- mice with spontaneous hyperlipidemia by feeding a normal diet, it significantly reduced the atherosclerotic lesion size by 40% in Hhcy-induced atherosclerosis without affecting plasma homocysteine levels and lipid contents, effectively suppressed PARP activation, and inhibited nuclear translocation of nuclear factor-(kappa)B (NF-(kappa)B) and subsequent production of inflam-matory factors, such as vascular cell adhesion molecule-1 (VCAM-1) and monocyte chemoattactant protein-1 (MCP-1). CONCLUSION: Our results suggest that PARP inhibition attenuates atherosclerotic plaque development under hyperhomocysteinemic conditions, through the inhibition of PARP activation, nuclear NF-kappaB translocation and subsequent expression of inflammatory factors.


Subject(s)
Apolipoproteins E/physiology , Atherosclerosis/prevention & control , Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Hyperhomocysteinemia/complications , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Apolipoproteins E/genetics , Apoptosis/drug effects , Atherosclerosis/complications , Base Sequence , Blotting, Western , DNA Primers , Enzyme Activation , Male , Mice , Mice, Knockout , NF-kappa B/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Polymerase Chain Reaction
15.
Cardiovasc Drugs Ther ; 23(2): 121-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18949543

ABSTRACT

INTRODUCTION: We investigated the possible protective effect of poly (ADP-ribose) polymerase (PARP) inhibition in preventing endothelial dysfunction induced by hyperhomocysteinemia (Hhcy). METHODS: Sprague-Dawley rats were divided into Hhcy group, Hhcy + 3-aminobenzamide(3-AB) group, control group and control + 3-AB group. A high-methionine diet was given to induce hyperhomocysteinemia. In Hhcy + 3-AB and control + 3-AB groups, rats were injected intraperitoneally with 3-AB (inhibitor of PARP). After 45 days, ultrastructural changes of aortas were observed by transmission electron microscope. Vascular reactivity of thoracic aortic rings was measured in organ chambers. PARP activity was detected. The levels of plasma total homocysteine, nitrite/nitrate, endothelin (ET)-1 and malondialdehyde were assayed. RESULTS: Rats in Hhcy group developed severe hyperhomocysteinemia and significant loss of endothelial function as measured by both vascular rings and levels of nitrite/nitrate and ET-1. Malondialdehyde levels increased significantly in Hhcy rats compared with control rats. 3-AB improved Ach-induced, NO-mediated vascular relaxation and stabilized the level of nitrite/nitrate and ET-1. Obvious improvement of ultrastructure can be observed in Hhcy + 3-AB group. CONCLUSIONS: These results suggest that pharmacological inhibition of PARP prevents the development of endothelial dysfunction in rats with hyperhomocysteinemia which may represent a novel approach to improve vascular dysfunction associated with hyperhomocysteinemia.


Subject(s)
Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Hyperhomocysteinemia/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Disease Models, Animal , Endothelin-1/drug effects , Endothelin-1/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Hyperhomocysteinemia/physiopathology , Male , Malondialdehyde/metabolism , Microscopy, Electron, Transmission , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
16.
J Mol Cell Cardiol ; 45(2): 168-75, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18502445

ABSTRACT

Atherosclerosis is a chronic inflammatory disease, which is positively and negatively regulated by T helper (Th) 1 and Th2 lymphocytes, respectively. Recent findings indicate that suppressive oligodeoxynucleotides (ODNs) expressing TTAGGG motifs selectively reduce Th1 cytokine production and have been proven effective at blocking the development of organ-specific autoimmune diseases. In the current research, we hypothesized that suppressive ODNs may alter the development of atherosclerosis. Eight-week-old homozygous ApoE(-/-) male mice were injected with 300 mug ODNs A151 (TTAGGG) or nonspecific ODNs 1612. Atherosclerotic lesion sizes were dramatically reduced by ODNs A151, but not by nonspecific ODNs. MCP-1 and VCAM-1, which are the key inflammatory factors in atherogenesis, were significantly attenuated by the suppressive ODNs A151. In the splenic lymphocytes, FACS analysis showed ODNs A151 reduced the percentage of IFN-gamma-producing Th1 cells and slightly increased the percentage of IL-4-producing Th2 cells, indicating that suppressive ODNs skewed the Th1/Th2 balance toward Th2 inflammation in vivo. Furthermore, ODNs A151 down-regulated the phosphorylation of STAT1 and STAT4 and suppressed up-regulation of T-bet, a signal modulator for Th1, and didn't impact GATA-3 and STAT6, which are associated with a Th2 phenotype. Consistent with this in vivo observation, ELISA analysis demonstrated that ODNs A151 suppressed Th1 cytokines IFN-gamma and TNF-alpha, and augmented Th2 cytokines IL-4 and IL-10 in vitro. This study provides the first experimental evidence that suppressive ODNs inhibit the development of atherosclerosis through inhibition of the STAT1/4 and T-bet pathways, which further modulate the Th1/Th2 balance in vivo.


Subject(s)
Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/therapy , Immunosuppressive Agents/administration & dosage , Oligodeoxyribonucleotides/administration & dosage , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/prevention & control , Cells, Cultured , CpG Islands/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology , Th1 Cells/drug effects , Th2 Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...