Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
J Mol Cell Cardiol ; 173: 30-46, 2022 12.
Article in English | MEDLINE | ID: mdl-36179399

ABSTRACT

Autophagy flux is impaired during myocardial ischemia/reperfusion (M-I/R) via the accumulation of autophagosome and insufficient clearance, which exacerbates cardiomyocyte death. Peli1 (Pellion1) is a RING finger domain-containing ubiquitin E3 ligase that could catalyze the polyubiquitination of substrate proteins. Peli1 has been demonstrated to play an important role in ischemic cardiac diseases. However, little is known about whether Peli1 is involved in the regulation of autophagy flux during M-I/R. The present study investigated whether M-I/R induced impaired autophagy flux could be mediated through Peli1 dependent mechanisms. We induced M-I/R injury in wild type (WT) and Peli1 knockout mice and observed that M-I/R significantly decreased cardiac function that was associated with increased cardiac Peli1 expression and upregulated autophagy-associated protein LC3II and P62. In contrast, Peli1 knockout mice exhibited significant improvement of M-I/R induced cardiac dysfunction and decreased LC3II and P62 expression. Besides, inhibitors of autophagy also increased the infarct size in Peli1 knockout mice after 24 h of reperfusion. Mechanistic studies demonstrated that in vivo I/R or in vitro hypoxia/reoxygenation (H/R) markedly increased the Peli1 E3 ligase activity which directly promoted the ubiquitination of P62 at lysine(K)7 via K63-linkage to inhibit its dimerization and autophagic degradation. Co-immunoprecipitation and GST-pull down assay indicated that Peli1 interacted with P62 via the Ring domain. In addition, Peli1 deficiency also decreased cardiomyocyte apoptosis. Together, our work demonstrated a critical link between increased expression and activity of Peli1 and autophagy flux blockage in M-I/R injury, providing insight into a promising strategy for treating myocardium M-I/R injury.


Subject(s)
Myocardial Reperfusion Injury , Mice , Animals , Myocardial Reperfusion Injury/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Autophagy , Myocytes, Cardiac/metabolism , Ubiquitination , Mice, Knockout , Nuclear Proteins/metabolism
2.
Basic Res Cardiol ; 115(2): 19, 2020 02 01.
Article in English | MEDLINE | ID: mdl-32008145

ABSTRACT

Activation of TLRs mediated the NF-κB signaling pathway plays an important pathophysiological role in cardiac hypertrophy. Triad3A, a ubiquitin E3 ligase, has been reported to negatively regulate NF-κB activation pathway via promoting ubiquitination and degradation of TLR4 and TLR9 in innate immune cells. The role of Triad3A in cardiac hypertrophic development remains unknown. The present study investigated whether there is a link between Triad3A and TLR4 and TLR9 in pressure overload induced cardiac hypertrophy. We observed that Triad3A levels were markedly reduced following transverse aortic constriction (TAC) induced cardiac hypertrophy. Similarly, stimulation of neonatal rat cardiac myocytes (NRCMs) with angiotensin-II (Ang II) significantly decreased Triad3A expression. To determine the role of Triad3A in TAC-induced cardiac hypertrophy, we transduced the myocardium with adenovirus expressing Triad3A followed by induction of TAC. We observed that increased expression of Triad3A significantly attenuated cardiac hypertrophy and improved cardiac function. To investigate the mechanisms by which Triad3A attenuated cardiac hypertrophy, we examined the Triad3A E3 ubiquitination on TLR4 and TLR9. We found that Triad3A promoted TLR4 and TLR9 degradation through ubiquitination. Triad3A mediated TLR4 and TLR9 degradation resulted in suppression of NF-κB activation. Our data suggest that Triad3A plays a protective role in the development of cardiac hypertrophy, at least through catalyzing ubiquitination-mediated degradation of TLR4 and TLR9, thus negatively regulating NF-κB activation.


Subject(s)
Hypertrophy, Left Ventricular/prevention & control , Myocardium/enzymology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism , Ubiquitin-Protein Ligases/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Cells, Cultured , Disease Models, Animal , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Male , Mice, Inbred C57BL , Myocardium/pathology , NF-kappa B/metabolism , Proteolysis , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
3.
J Infect Dis ; 215(9): 1396-1406, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28368517

ABSTRACT

Background: Cardiac dysfunction is present in >40% of sepsis patients and is associated with mortality rates of up to 70%. Recent evidence suggests that glycolytic metabolism plays a critical role in host defense and inflammation. Activation of Toll-like receptors on immune cells can enhance glycolytic metabolism. This study investigated whether modulation of glycolysis by inhibition of hexokinase will be beneficial to septic cardiomyopathy. Methods: Male C57B6/J mice were treated with a hexokinase inhibitor (2-deoxy-d-glucose [2-DG], 0.25-2 g/kg, n = 6-8) before cecal ligation and puncture (CLP) induced sepsis. Untreated septic mice served as control. Sham surgically operated mice treated with or without the 2-DG inhibitor served as sham controls. Cardiac function was assessed 6 hours after CLP sepsis by echocardiography. Serum was harvested for measurement of inflammatory cytokines and lactate. Results: Sepsis-induced cardiac dysfunction was significantly attenuated by administration of 2-DG. Ejection fraction and fractional shortening in 2-DG-treated septic mice were significantly (P < .05) greater than in untreated CLP mice. 2-DG administration also significantly improved survival outcome, reduced kidney and liver injury, attenuated sepsis-increased serum levels of tumor necrosis factor α and interleukin 1ß as well as lactate, and enhanced the expression of Sirt1 and Sirt3 in the myocardium, which play an important role in mitochondrial function and metabolism. In addition, 2-DG administration suppresses sepsis-increased expression of apoptotic inducers Bak and Bax as well as JNK phosphorylation in the myocardium. Conclusions: Glycolytic metabolism plays an important role in mediating sepsis-induced septic cardiomyopathy. The mechanisms may involve regulation of inflammatory response and apoptotic signaling.


Subject(s)
Cardiomyopathies/metabolism , Glycolysis/physiology , Heart/physiopathology , Sepsis/metabolism , Animals , Cardiomyopathies/physiopathology , Cytokines/metabolism , Deoxyglucose/metabolism , Deoxyglucose/pharmacology , Deoxyglucose/therapeutic use , Disease Models, Animal , Glycolysis/drug effects , Heart/drug effects , Hexokinase/antagonists & inhibitors , Hexokinase/metabolism , Lactic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Sepsis/drug therapy , Sepsis/mortality , Sepsis/physiopathology , Survival Analysis
4.
Biochim Biophys Acta ; 1862(6): 1191-202, 2016 06.
Article in English | MEDLINE | ID: mdl-26964796

ABSTRACT

The TIR/BB-loop mimetic AS-1 has been reported to prevent cardiac hypertrophy by inhibiting interleukin-1 receptor (IL-1R)-mediated myeloid differentiation primary response gene 88 (MyD88)-dependent signaling. To date, it remains unknown whether and if so how AS-1 contributes to mechanical stress (MS)-induced cardiac fibroblast activation, a key process in pressure overload-induced cardiac remodeling and heart failure. Here, we show that phosphorylation and expression of large tumor suppressor kinase 1 (LATS1), a key molecule in the Hippo-Yes associated protein (YAP) signaling pathway, were down-regulated in primary neonatal rat cardiac fibroblasts (NRCFs) in response to MS and in the hearts of mice subjected to transverse aortic constriction (TAC) procedure; AS-1 treatment was able to restore LATS1 phosphorylation and expression both in vitro and in vivo. AS-1 treatment suppressed the induction of proliferation, differentiation and collagen synthesis in response to MS in NRCFs. AS-1 also ameliorated cardiomyocyte hypertrophy and apoptosis through dampening paracrine secretion of stretched cardiac fibroblasts. In mice, AS-1 treatment could protect against TAC-induced cardiac hypertrophy, myocardial fibrosis and heart failure. Of note, LATS1 depletion using siRNA completely abrogated the inhibitory effects of AS-1 on NRCFs under MS including accelerated proliferation, differentiation, enhanced ability to produce collagen and augmented paracrine secretion of tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) to induce cardiomyocyte hypertrophy. Therefore, our results delineate a previously unrecognized role for LATS1 in cardiac fibroblast to mediate the beneficial effects of AS-1 in preventing pressure overload-induced cardiac remodeling and heart failure.


Subject(s)
Cardiomegaly/drug therapy , Cardiotonic Agents/therapeutic use , Fibroblasts/drug effects , Paracrine Communication/drug effects , Protein Serine-Threonine Kinases/metabolism , Pyrrolidines/therapeutic use , Animals , Biomimetic Materials/therapeutic use , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Male , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Stress, Mechanical
5.
J Immunol ; 195(2): 672-82, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26048146

ABSTRACT

Cardiac dysfunction is a major consequence of sepsis/septic shock and contributes to the high mortality of sepsis. Innate and inflammatory responses mediated by TLRs play a critical role in sepsis-induced cardiac dysfunction. MicroRNA-146 (miR-146) was first identified as a negative regulator in innate immune and inflammatory responses induced by LPS. This study examined whether miR-146a will have a protective effect on sepsis-induced cardiac dysfunction. Lentivirus-expressing miR-146a (LmiR-146a) or lentivirus-expressing scrambled miR (LmiR-control) was delivered into the myocardium via the right carotid artery. Seven days after transfection, mice were subjected to cecal ligation and puncture (CLP). Untransfected mice were also subjected to CLP-induced sepsis. Cardiac function was examined by echocardiography before and 6 h after CLP. In vitro studies showed that increased miR-146a levels suppress LPS-induced IκBα phosphorylation and inflammatory cytokine production in both H9C2 cardiomyocytes and J774 macrophages. In vivo transfection of LmiR-146a attenuated sepsis-induced cardiac dysfunction. The values for percent ejection fraction and percent fractional shortening in LmiR-146a-transfected CLP mice were significantly greater than in untransfected CLP control. LmiR-146a transfection prevented sepsis-induced NF-κB activity, suppressed IRAK and TRAF6 expression in the myocardium, and attenuated sepsis-induced inflammatory cytokine production in both plasma and peritoneal fluid. In addition, LmiR-146a transfection decreased sepsis-induced infiltration of neutrophils and macrophages into the myocardium. LmiR-146a can also transfect macrophages in the periphery. We conclude that miR-146a attenuates sepsis-induced cardiac dysfunction by preventing NF-κB activation, inflammatory cell infiltration, and inflammatory cytokine production via targeting of IRAK and TRAF6 in both cardiomyocytes and inflammatory monocytic cells.


Subject(s)
Heart Failure/therapy , Interleukin-1 Receptor-Associated Kinases/immunology , MicroRNAs/immunology , NF-kappa B/immunology , Sepsis/therapy , TNF Receptor-Associated Factor 6/immunology , Administration, Intravenous , Animals , Carotid Arteries , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Gene Expression Regulation , Genetic Vectors/administration & dosage , Heart Failure/etiology , Heart Failure/genetics , Heart Failure/immunology , Immunity, Innate , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/genetics , Lentivirus/genetics , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Myocardium/immunology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Primary Cell Culture , Sepsis/complications , Sepsis/genetics , Sepsis/immunology , Signal Transduction , TNF Receptor-Associated Factor 6/antagonists & inhibitors , TNF Receptor-Associated Factor 6/genetics
6.
J Infect Dis ; 214(11): 1773-1783, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27683819

ABSTRACT

BACKGROUND: This study examined the effect of microRNA-125b (miR-125b) on sepsis-induced cardiac dysfunction. METHODS: Mouse hearts were transfected with lentivirus expressing miR-125b (LmiR-125b) 7 days before cecal ligation and puncture (CLP)-induced sepsis. Cardiac function was examined by echocardiography before and 6 hours after CLP (n = 6/group). Survival was monitored following CLP-induced sepsis (n = 12/group). RESULTS: LmiR-125b transfection significantly attenuated cardiac dysfunction due to CLP-induced sepsis. Fractional shortening and ejection fraction values were significantly (P < .05) higher in the LmiR-125b-treated CLP group than in the untreated CLP group. Survival outcome in LmiR-125b-transfected septic mice was markedly improved, compared with mice with CLP-induced sepsis. Transfection of LmiR-125b into the heart significantly suppressed the expression of ICAM-1 and VCAM-1, decreased the accumulation of macrophages and neutrophils in the myocardium, and decreased serum levels of tumor necrosis factor α and interleukin 1ß by targeting tumor necrosis factor receptor-associated factor 6 (TRAF6)-mediated nuclear factor κB (NF-κB) activation. In addition, sepsis-induced myocardial apoptosis was markedly attenuated by LmiR-125b transfection through suppression of p53, Bax, and Bak1 expression. In vitro transfection of endothelial cells with miR-125b mimics attenuate LPS-induced ICAM-1 and VCAM-1 expression by suppressing TRAF6 and NF-κB activation. CONCLUSIONS: Increased myocardial miR-125b expression attenuates sepsis-induced cardiac dysfunction and improves survival. miR-125b may be a target for septic cardiomyopathy.


Subject(s)
Coinfection/pathology , Heart Failure/prevention & control , MicroRNAs/metabolism , NF-kappa B/metabolism , Sepsis/pathology , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism , Animals , Coinfection/complications , Disease Models, Animal , Echocardiography , Heart Failure/diagnostic imaging , Male , Mice , Mice, Inbred C57BL , Myocardium/pathology , Sepsis/complications , Survival Analysis
7.
J Mol Cell Cardiol ; 89(Pt A): 87-97, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26458524

ABSTRACT

OBJECTIVE: Activation of PI3K/Akt signaling protects the myocardium from ischemia/reperfusion injury. MicroRNAs have been demonstrated to play an important role in the regulation of gene expression at the post-transcriptional level. In this study, we examined whether miR-130a will attenuate cardiac dysfunction and remodeling after myocardial infarction (MI) via PI3K/Akt dependent mechanism. APPROACHES AND RESULTS: To determine the role of miR-130a in the proliferation and migration of endothelial cells, HUVECs were transfected with miR-130a mimics before the cells were subjected to scratch-induced wound injury. Transfection of miR-130a mimics stimulated the migration of endothelial cells into the wound area and increased phospho-Akt levels. To examine the effect of miR-130a on cardiac dysfunction and remodeling after MI, Lentivirus expressing miR-130a (LmiR-130a) was delivered into mouse hearts seven days before the mice were subjected to MI. Cardiac function was assessed by echocardiography before and for up to 21 days after MI. Ejection fraction (EF%) and fractional shortening (FS%) in the LmiR-130a transfected MI hearts were significantly greater than in LmiR-control and untransfected control MI groups. LmiR-130a transfection increased capillary number and VEGF expression, and decreased collagen deposition in the infarcted myocardium. Importantly, LmiR-130a transfection significantly suppressed PTEN expression and increased the levels of phosphorylated Akt in the myocardium. However, treatment of LmiR-130a-transfected mice with LY294002, a PI3K inhibitor, completely abolished miR-130a-induced attenuation of cardiac dysfunction after MI. CONCLUSIONS: miR-130a plays a critical role in attenuation of cardiac dysfunction and remodeling after MI. The mechanisms involve activation of PI3K/Akt signaling via suppression of PTEN expression.


Subject(s)
Heart/physiopathology , MicroRNAs/metabolism , Myocardial Infarction/physiopathology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Animals , Apoptosis , Cardiotonic Agents/metabolism , Cell Movement , Collagen/metabolism , Enzyme Activation , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Lentivirus/metabolism , Ligands , Male , Matrix Metalloproteinase 2/metabolism , Mice, Inbred C57BL , Microvessels/pathology , Myocardial Infarction/complications , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocardium/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Toll-Like Receptors/metabolism , Transfection , Vascular Endothelial Growth Factor A/metabolism
8.
J Mol Cell Cardiol ; 79: 145-56, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25446187

ABSTRACT

Activation of cardiac fibroblasts is a key event in the progression of cardiac fibrosis that leads to heart failure. However, the molecular mechanisms underlying mechanical stress-induced cardiac fibroblast activation are complex and poorly understood. This study demonstrates that Pellino1, an E3 ubiquitin ligase, was activated in vivo in pressure overloaded rat hearts and in cultured neonatal rat cardiac fibroblasts (NRCFs) exposed to mechanical stretch in vitro. Suppression of the expression and activity of Pellino1 by adenovirus-mediated delivery of shPellino1 (adv-shpeli1) attenuated pressure overload-induced cardiac dysfunction and cardiac hypertrophy and decreased cardiac fibrosis in rat hearts. Transfection of adv-shpeli1 also significantly attenuated mechanical stress-induced proliferation, differentiation and collagen synthesis in NRCFs. Pellino1 silencing also abrogated mechanical stretch-induced polyubiquitination of tumor necrosis factor-alpha receptor association factor-6 (TRAF6) and receptor-interacting protein 1 (RIP1) and consequently decreased the DNA binding activity of nuclear factor-kappa B (NF-κB) in NRCFs. In addition, Pellino1 silencing prevented stretch-induced activation of p38 and activator protein 1 (AP-1) binding activity in NRCFs. Chromatin Immunoprecipitation (ChIP) and luciferase reporter assays showed that Pellino1 silencing prevented the binding of NF-κB and AP-1 to the promoter region of transforming growth factor-ß1 (TGF-ß1) thus dampening TGF-ß1 transactivation. Our data reveal a previously unrecognized role of Pellino1 in extracellular matrix deposition and cardiac fibroblast activation in response to mechanical stress and provides a novel target for treatment of cardiac fibrosis and heart failure.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/pathology , Myocardium/pathology , Nuclear Proteins/metabolism , Stress, Mechanical , Transforming Growth Factor beta1/biosynthesis , Adenoviridae/metabolism , Animals , Animals, Newborn , Cell Differentiation , Collagen/metabolism , Fibrosis , Gene Silencing , Heart/physiopathology , Male , Models, Biological , Myocardium/metabolism , NF-kappa B/metabolism , Nuclear Proteins/antagonists & inhibitors , Phosphorylation , Polyubiquitin/metabolism , Pressure , Promoter Regions, Genetic/genetics , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinases , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta1/genetics , Ubiquitin-Protein Ligases , Ubiquitination , p38 Mitogen-Activated Protein Kinases/metabolism
9.
J Cell Mol Med ; 19(3): 555-65, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25351293

ABSTRACT

Toll-like receptor (TLR)-mediated signalling plays a role in cerebral ischaemia/reperfusion (I/R) injury. Modulation of TLRs has been reported to protect against cerebral I/R injury. This study examined whether modulation of TLR3 with poly (I:C) will induce protection against cerebral I/R injury. Mice were treated with or without Poly (I:C) (n = 8/group) 1 hr prior to cerebral ischaemia (60 min.) followed by reperfusion (24 hrs). Poly (I:C) pre-treatment significantly reduced the infarct volume by 57.2% compared with untreated I/R mice. Therapeutic administration of Poly (I:C), administered 30 min. after cerebral ischaemia, markedly decreased infarct volume by 34.9%. However, Poly (I:C)-induced protection was lost in TLR3 knockout mice. In poly (I:C)-treated mice, there was less neuronal damage in the hippocampus compared with untreated I/R mice. Poly (I:C) treatment induced IRF3 phosphorylation, but it inhibited NF-κB activation in the brain. Poly (I:C) also decreased I/R-induced apoptosis by attenuation of Fas/FasL-mediated apoptotic signalling. In addition, Poly (I:C) treatment decreased microglial cell caspase-3 activity. In vitro data showed that Poly (I:C) prevented hypoxia/reoxygenation (H/R)-induced interaction between Fas and FADD as well as caspase-3 and -8 activation in microglial cells. Importantly, Poly (I:C) treatment induced co-association between TLR3 and Fas. Our data suggest that Poly (I:C) decreases in cerebral I/R injury via TLR3 which associates with Fas, thereby preventing the interaction of Fas and FADD, as well as microglial cell caspase-3 and -8 activities. We conclude that TLR3 modulation by Poly (I:C) could be a potential approach for protection against ischaemic stroke.


Subject(s)
Cerebral Infarction/drug therapy , Fas-Associated Death Domain Protein/metabolism , Poly I-C/therapeutic use , Toll-Like Receptor 3/metabolism , fas Receptor/metabolism , Animals , Apoptosis/drug effects , Brain/blood supply , Brain/physiopathology , Caspase 3/biosynthesis , Caspase 3/metabolism , Caspase 8/biosynthesis , Caspase 8/metabolism , Cell Hypoxia/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Neuroprotective Agents/therapeutic use , Reperfusion Injury/drug therapy , Toll-Like Receptor 3/biosynthesis , Toll-Like Receptor 3/genetics
10.
Biochim Biophys Acta ; 1842(1): 22-31, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24140513

ABSTRACT

Innate immune and inflammatory responses mediated by Toll like receptors (TLRs) have been implicated in myocardial ischemia/reperfusion (I/R) injury. This study examined the role of TLR3 in myocardial injury induced by two models, namely, myocardial infarction (MI) and I/R. First, we examined the role of TLR3 in MI. TLR3 deficient (TLR3(-/-)) and wild type (WT) mice were subjected to MI induced by permanent ligation of the left anterior descending (LAD) coronary artery for 21days. Cardiac function was measured by echocardiography. Next, we examined whether TLR3 contributes to myocardial I/R injury. TLR3(-/-) and WT mice were subjected to myocardial ischemia (45min) followed by reperfusion for up to 3days. Cardiac function and myocardial infarct size were examined. We also examined the effect of TLR3 deficiency on I/R-induced myocardial apoptosis and inflammatory cytokine production. TLR3(-/-) mice showed significant attenuation of cardiac dysfunction after MI or I/R. Myocardial infarct size and myocardial apoptosis induced by I/R injury were significantly attenuated in TLR3(-/-) mice. TLR3 deficiency increases B-cell lymphoma 2 (BCL2) levels and attenuates I/R-increased Fas, Fas ligand or CD95L (FasL), Fas-Associated protein with Death Domain (FADD), Bax and Bak levels in the myocardium. TLR3 deficiency also attenuates I/R-induced myocardial nuclear factor KappaB (NF-κB) binding activity, Tumor necrosis factor alpha (TNF-α) and Interleukin-1 beta (IL-1ß) production as well as I/R-induced infiltration of neutrophils and macrophages into the myocardium. TLR3 plays an important role in myocardial injury induced by MI or I/R. The mechanisms involve activation of apoptotic signaling and NF-κB binding activity. Modulation of TLR3 may be an effective approach for ameliorating heart injury in heart attack patients.


Subject(s)
Gene Expression Regulation , Myocardial Infarction/genetics , Myocardial Reperfusion Injury/genetics , Toll-Like Receptor 3/genetics , Animals , Apoptosis , Disease Models, Animal , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Neutrophils/metabolism , Neutrophils/pathology , Severity of Illness Index , Signal Transduction , Toll-Like Receptor 3/deficiency , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , fas Receptor/genetics , fas Receptor/metabolism
11.
Biochim Biophys Acta ; 1832(2): 336-46, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23123599

ABSTRACT

The macrophage scavenger receptor class A (SR-A) participates in the innate immune and inflammatory responses. This study examined the role of macrophage SR-A in myocardial ischemia/reperfusion (I/R) injury and hypoxia/reoxygenation (H/R)-induced cell damage. SR-A(-/-) and WT mice were subjected to ischemia (45min) followed by reperfusion for up to 7days. SR-A(-/-) mice showed smaller myocardial infarct size and better cardiac function than did WT I/R mice. SR-A deficiency attenuated I/R-induced myocardial apoptosis by preventing p53-mediated Bak-1 apoptotic signaling. The levels of microRNA-125b in SR-A(-/-) heart were significantly greater than in WT myocardium. SR-A is predominantly expressed on macrophages. To investigate the role of SR-A macrophages in H/R-induced injury, we isolated peritoneal macrophages from SR-A deficient (SR-A(-/-)) and wild type (WT) mice. Macrophages were subjected to hypoxia followed by reoxygenation. H/R markedly increased NF-κB binding activity as well as KC and MCP-1 production in WT macrophages but not in SR-A(-/-) macrophages. H/R induced caspase-3/7 and -8 activities and cell death in WT macrophages, but not in SR-A(-/-) macrophages. The levels of miR-125b in SR-A(-/-) macrophages were significantly higher than in WT macrophages. Transfection of WT macrophages with miR-125b mimics attenuated H/R-induced caspase-3/7 and -8 activities and H/R-decreased viability, and prevented H/R-increased p-53, Bak-1 and Bax expression. The data suggest that SR-A deficiency attenuates myocardial I/R injury by targeting p53-mediated apoptotic signaling. SR-A(-/-) macrophages contain high levels of miR-125b which may play a role in the protective effect of SR-A deficiency on myocardial I/R injury and H/R-induced cell damage.


Subject(s)
Macrophages/metabolism , MicroRNAs/genetics , Myocardial Reperfusion Injury/genetics , Scavenger Receptors, Class A/physiology , Animals , Apoptosis , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Scavenger Receptors, Class A/genetics
12.
Biochim Biophys Acta ; 1832(1): 96-104, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22917564

ABSTRACT

BACKGROUND: Toll-like receptors (TLRs) have been implicated in myocardial ischemia/reperfusion (I/R) injury. The TLR9 ligand, CpG-ODN has been reported to improve cell survival. We examined effect of CpG-ODN on myocardial I/R injury. METHODS: Male C57BL/6 mice were treated with either CpG-ODN, control-ODN, or inhibitory CpG-ODN (iCpG-ODN) 1h prior to myocardial ischemia (60min) followed by reperfusion. Untreated mice served as I/R control (n=10/each group). Infarct size was determined by TTC straining. Cardiac function was examined by echocardiography before and after myocardial I/R up to 14days. RESULTS: CpG-ODN administration significantly decreased infarct size by 31.4% and improved cardiac function after myocardial I/R up to 14days. Neither control-ODN nor iCpG-ODN altered I/R-induced myocardial infarction and cardiac dysfunction. CpG-ODN attenuated I/R-induced myocardial apoptosis and prevented I/R-induced decrease in Bcl2 and increase in Bax levels in the myocardium. CpG-ODN increased Akt and GSK-3ß phosphorylation in the myocardium. In vitro data suggested that CpG-ODN treatment induced TLR9 tyrosine phosphorylation and promoted an association between TLR9 and the p85 subunit of PI3K. Importantly, PI3K/Akt inhibition and Akt kinase deficiency abolished CpG-ODN-induced cardioprotection. CONCLUSION: CpG-ODN, the TLR9 ligand, induces protection against myocardial I/R injury. The mechanisms involve activation of the PI3K/Akt signaling pathway.


Subject(s)
Myocardial Ischemia/surgery , Oligodeoxyribonucleotides/administration & dosage , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Toll-Like Receptor 9/agonists , Animals , Cell Line , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats , Reperfusion Injury/genetics , Signal Transduction/drug effects , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism
13.
PLoS Pathog ; 8(10): e1002967, 2012.
Article in English | MEDLINE | ID: mdl-23071440

ABSTRACT

Sepsis is a frequent complication in critical illness. The mechanisms that are involved in initiation and propagation of the disease are not well understood. Scavenger receptor A (SRA) is a membrane receptor that binds multiple polyanions such as oxidized LDL and endotoxin. Recent studies suggest that SRA acts as a pattern recognition receptor in the innate immune response. The goal of the present study was to determine the role of SRA in polymicrobial sepsis. SRA deficient (SRA(-/-)) and C57BL/6JB/6J (WT) male mice were subjected to cecal ligation and puncture (CLP) to induce polymicrobial sepsis. NFκB activity, myeloperoxidase activity, and co-association of SRA with toll like receptor (TLR) 4 and TLR2 was analyzed in the lungs. Spleens were analyzed for apoptosis. Serum cytokines and chemokines were assayed. Blood and peritoneal fluid were cultured for aerobic and anaerobic bacterial burdens. Long-term survival was significantly increased in SRA(-/-) septic mice (53.6% vs. 3.6%, p < 0.05) when compared to WT mice. NFκB activity was 45.5% lower in the lungs of SRA(-/-) septic mice versus WT septic mice (p < 0.05). Serum levels of interleukin (IL)-5, IL-6, IL-10 and monocyte chemoattractant protein -1 were significantly lower in septic SRA(-/-) mice when compared to septic WT mice (p < 0.05). We found that SRA immuno-precipitated with TLR4, but not TLR2, in the lungs of WT septic mice. We also found that septic SRA(-/-) mice had lower bacterial burdens than WT septic mice. SRA deficiency had no effect on pulmonary neutrophil infiltration or splenocyte apoptosis during sepsis. We conclude that SRA plays a pivotal, and previously unknown, role in mediating the pathophysiology of sepsis/septic shock in a murine model of polymicrobial sepsis. Mechanistically, SRA interacts with TLR4 to enhance the development of the pro-inflammatory phenotype and mediate the morbidity and mortality of sepsis/septic shock.


Subject(s)
Coinfection/immunology , Scavenger Receptors, Class A/metabolism , Sepsis/immunology , Toll-Like Receptor 4/metabolism , Animals , Apoptosis , Ascitic Fluid/microbiology , Bacterial Load , Blood/microbiology , Cecum/surgery , Chemokines/blood , Chemokines/immunology , Coinfection/microbiology , Coinfection/mortality , Cytokines/blood , Cytokines/immunology , Gene Expression Regulation , Lung/immunology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Neutrophil Infiltration , Peroxidase , Scavenger Receptors, Class A/deficiency , Scavenger Receptors, Class A/genetics , Sepsis/microbiology , Sepsis/mortality , Shock, Septic/microbiology , Spleen/immunology , Spleen/metabolism , Toll-Like Receptor 2/metabolism
14.
J Infect Dis ; 207(9): 1471-9, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23359590

ABSTRACT

BACKGROUND: Toll-like receptors (TLRs) play a role in the pathophysiology of sepsis and multiple organ failure. This study examined the effect of CpG oligodeoxynucleotide (CpG-ODN), the TLR9 ligand, on polymicrobial sepsis-induced cardiac dysfunction. METHODS: Male C57BL/6 mice were treated with CpG-ODN, control CpG-ODN (control-ODN), or inhibitory CpG-ODN (iCpG-ODN) 1 hour prior to cecal ligation and puncture (CLP)-induced sepsis. Mice that underwent sham surgery served as sham controls. Cardiac function was examined by echocardiography before and 6 hours after CLP. RESULTS: Cardiac function was significantly decreased 6 hours after CLP. CpG-ODN prevented CLP-induced cardiac dysfunction, as evidenced by maintenance of the ejection fraction and fractional shortening. Control-ODN or iCpG-ODN did not alter CLP-induced cardiac dysfunction. CpG-ODN significantly attenuated CLP-induced myocardial apoptosis and increased myocardial Akt and extracellular-signal-related kinase (ERK) phosphorylation levels following CLP. In vitro experiments demonstrated that CpG-ODN promotes an association between TLR9 and Ras, resulting in Akt and ERK phosphorylation. Inhibition of phosphoinositide 3-kinase (PI3K) by Ly294002 or inhibition of ERK by U0126 in vivo abolished CpG-ODN attenuation of CLP-induced cardiac dysfunction. CONCLUSIONS: CpG-ODN prevents CLP-induced cardiac dysfunction, in part through activation of PI3K/Akt and ERK signaling. Modulation of TLR9 could be an effective approach for treatment of cardiovascular dysfunction in patients with sepsis or septic shock.


Subject(s)
Cardiovascular Agents/administration & dosage , Heart Failure/prevention & control , Heart/drug effects , Heart/physiopathology , Oligodeoxyribonucleotides/administration & dosage , Sepsis/complications , Sepsis/drug therapy , Animals , Echocardiography , Male , Mice , Mice, Inbred C57BL , Treatment Outcome
15.
Biochim Biophys Acta ; 1823(7): 1192-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22627090

ABSTRACT

Recent evidence suggests that the macrophage scavenger receptor class A (SR-A, aka, CD204) plays a role in the induction of innate immune and inflammatory responses. We investigated whether SR-A will cooperate with Toll-like receptors (TLRs) in response to TLR ligand stimulation. Macrophages (J774/a) were treated with Pam2CSK4, (TLR2 ligand), Polyinosinic:polycytidylic acid (Poly I:C) (TLR3 ligand), and Lipopolysaccharides (LPS) (TLR4 ligand) for 15 min in the presence or absence of fucoidan (the SR-A ligand). The levels of phosphorylated IκBα (p-IκBα) were examined by Western blot. We observed that Poly I:C and LPS alone, but not Pam2CSK4 or fucoidan increased the levels of p-IκBα. However, LPS-induced increases in p-IκBα levels were further enhanced when presence of the fucoidan. Immunoprecipitation and double fluorescent staining showed that LPS stimulation promotes SR-A association with TLR4 in the presence of fucoidan. To further confirm our observation, we isolated peritoneal macrophages from SR-A deficient (SR-A(-/-)), TLR4(-/-) and wild type (WT) mice, respectively. The peritoneal macrophages were treated with LPS for 15min in the presence and absence of fucoidan. We observed that LPS-stimulated TNFα and IL-1ß production was further enhanced in the WT macrophages, but did not in either TLR4(-/-) or SR-A(-/-) macrophages, when fucoidan was present. Similarly, in the presence of fucoidan, LPS-induced IκBα phosphorylation, NF-κB binding activity, and association between TLR4 and SR-A were significantly enhanced in WT macrophages compared with LPS stimulation alone. The data suggests that SR-A is needed for LPS-induced inflammatory responses in macrophages.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , NF-kappa B/metabolism , Scavenger Receptors, Class A/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cytokines/metabolism , Drug Synergism , I-kappa B Proteins/metabolism , Inflammation Mediators/metabolism , Ligands , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , Phosphorylation/drug effects , Polysaccharides/pharmacology , Protein Binding/drug effects , Time Factors
16.
J Immunol ; 187(3): 1458-66, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21709150

ABSTRACT

This study examined the effect of TLR2 activation by its specific ligand, Pam3CSK4, on cerebral ischemia/reperfusion (I/R) injury. Mice (n = 8/group) were treated with Pam3CSK4 1 h before cerebral ischemia (60 min), followed by reperfusion (24 h). Pam3CSK4 was also given to the mice (n = 8) 30 min after ischemia. Infarct size was determined by triphenyltetrazolium chloride staining. The morphology of neurons in brain sections was examined by Nissl staining. Pam3CSK4 administration significantly reduced infarct size by 55.9% (p < 0.01) compared with untreated I/R mice. Therapeutic treatment with Pam3CSK4 also significantly reduced infarct size by 55.8%. Morphologic examination showed that there was less neuronal damage in the hippocampus of Pam3CSK4-treated mice compared with untreated cerebral I/R mice. Pam3CSK4 treatment increased the levels of Hsp27, Hsp70, and Bcl2, and decreased Bax levels and NF-κB-binding activity in the brain tissues. Administration of Pam3CSK4 significantly increased the levels of phospho-Akt/Akt and phospho-GSK-3ß/GSK-3ß compared with untreated I/R mice. More significantly, either TLR2 deficiency or PI3K inhibition with LY29004 abolished the protection by Pam3CSK4. These data demonstrate that activation of TLR2 by its ligand prevents focal cerebral ischemic damage through a TLR2/PI3K/Akt-dependent mechanism. Of greater significance, these data indicate that therapy with a TLR2-specific agonist during cerebral ischemia is effective in reducing injury.


Subject(s)
Infarction, Middle Cerebral Artery/immunology , Lipopeptides/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/physiology , Reperfusion Injury/immunology , Signal Transduction/immunology , Toll-Like Receptor 2/agonists , Animals , Enzyme Activation/immunology , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/prevention & control , Ligands , Lipopeptides/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphatidylinositol 3-Kinase/physiology , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/enzymology , Reperfusion Injury/prevention & control , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/metabolism
17.
Sci Adv ; 9(5): eadc9465, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36735787

ABSTRACT

High levels of lactate are positively associated with the prognosis and mortality in patients with heart attack. Endothelial-to-mesenchymal transition (EndoMT) plays an important role in cardiac fibrosis. Here, we report that lactate exerts a previously unknown function that increases cardiac fibrosis and exacerbates cardiac dysfunction by promoting EndoMT following myocardial infarction (MI). Treatment of endothelial cells with lactate disrupts endothelial cell function and induces mesenchymal-like function following hypoxia by activating the TGF-ß/Smad2 pathway. Mechanistically, lactate induces an association between CBP/p300 and Snail1, leading to lactylation of Snail1, a TGF-ß transcription factor, through lactate transporter monocarboxylate transporter (MCT)-dependent signaling. Inhibiting Snail1 diminishes lactate-induced EndoMT and TGF-ß/Smad2 activation after hypoxia/MI. The MCT inhibitor CHC mitigates lactate-induced EndoMT and Snail1 lactylation. Silence of MCT1 compromises lactate-promoted cardiac dysfunction and EndoMT after MI. We conclude that lactate acts as an important molecule that up-regulates cardiac EndoMT after MI via induction of Snail1 lactylation.


Subject(s)
Endothelial Cells , Myocardial Infarction , Humans , Endothelial Cells/metabolism , Lactic Acid , Transforming Growth Factor beta/metabolism , Myocardial Infarction/metabolism , Hypoxia/metabolism , Fibrosis
18.
J Leukoc Biol ; 113(2): 95-108, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36822176

ABSTRACT

The polarization of macrophages to the M1 or M2 phenotype has a pivotal role in inflammatory response following myocardial ischemia/reperfusion injury. Peli1, an E3 ubiquitin ligase, is closely associated with inflammation and autoimmunity as an important regulatory protein in the Toll-like receptor signaling pathway. We aimed to explore the function of Peli1 in macrophage polarization under myocardial ischemia/reperfusion injury and elucidate the possible mechanisms. We show here that Peli1 is upregulated in peripheral blood mononuclear cells from patients with myocardial ischemia/reperfusion, which is correlated with myocardial injury and cardiac dysfunction. We also found that the proportion of M1 macrophages was reduced and myocardial infarct size was decreased, paralleling improvement of cardiac function in mice with Peli1 deletion in hematopoietic cells or macrophages. Macrophage Peli1 deletion lessened M1 polarization and reduced the migratory ability in vitro. Mechanistically, Peli1 contributed to M1 polarization by promoting K63-linked ubiquitination and nuclear translocation of IRF5. Moreover, Peli1 deficiency in macrophages reduced the apoptosis of cardiomyocytes in vivo and in vitro. Together, our study demonstrates that Peli1 deficiency in macrophages suppresses macrophage M1 polarization and alleviates myocardial ischemia/reperfusion injury by inhibiting the nuclear translocation of IRF5, which may serve as a potential intervention target for myocardial ischemia/reperfusion injury.


Subject(s)
Myocardial Reperfusion Injury , Reperfusion Injury , Mice , Animals , Myocardial Reperfusion Injury/metabolism , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , Signal Transduction , Interferon Regulatory Factors/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
19.
Crit Care Med ; 40(8): 2390-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22635047

ABSTRACT

OBJECTIVE: To determine the role of Toll-like receptor 3 in cardiac dysfunction during polymicrobial sepsis. DESIGN: Controlled animal study. SETTING: University research laboratory. SUBJECTS: Male C57BL/6, wild-type, Toll-like receptor 3-/-. INTERVENTION: Myocardial dysfunction is a major consequence of septic shock and contributes to the high mortality of sepsis. Toll-like receptors (TLRs) play a critical role in the pathophysiology of sepsis/septic shock. TLR3 is located in intracellular endosomes, and recognizes double-stranded RNA. This study examined the role of TLR3 in cardiac dysfunction following cecal ligation and puncture (CLP)-induced sepsis. TLR3 knockout (TLR3-/-, n=12) and age-matched wild-type (n=12) mice were subjected to CLP. Cardiac function was measured by echocardiography before and 6 hrs after CLP. MEASUREMENTS AND MAIN RESULTS: CLP resulted in significant cardiac dysfunction as evidenced by decreased ejection fraction by 25.7% and fractional shortening by 29.8%, respectively. However, TLR3-/- mice showed a maintenance of cardiac function at pre-CLP levels. Wild-type mice showed 50% mortality at 58 hrs and 100% mortality at 154 hrs after CLP. In striking contrast, 70% of TLR3-/- mice survived indefinitely, that is, >200 hrs. TLR3 deficiency significantly decreased CLP-induced cardiac-myocyte apoptosis and attenuated CLP-induced Fas and Fas ligand expression in the myocardium. CLP-activation of TLR4-mediated nuclear factor-κB and Toll/IL-1 receptor-domain-containing adapter-inducing interferon-ß-dependant interferon signaling pathways was prevented by TLR3 deficiency. In addition, CLP-increased vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 expression, and neutrophil and macrophage sequestration in the myocardium were also attenuated in septic TLR3-/- mice. More significantly, adoptive transfer of wild-type bone-marrow stromal cells to TLR3-/- mice abolished the cardioprotective effect in sepsis. CONCLUSIONS: These data indicate that TLR3 plays a deleterious role in mediating cardiac dysfunction in sepsis. Thus, modulation of the TLR3 activity may be useful in preventing cardiac dysfunction in sepsis.


Subject(s)
Heart/physiopathology , Sepsis/physiopathology , Toll-Like Receptor 3/physiology , Animals , Apoptosis/physiology , Blotting, Western , Echocardiography , Electrophoretic Mobility Shift Assay , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils/physiology , Peroxidase/metabolism , Sepsis/microbiology
20.
Clin Mol Allergy ; 10: 3, 2012 Feb 06.
Article in English | MEDLINE | ID: mdl-22309647

ABSTRACT

BACKGROUND: Human mast cells are capable of a wide variety of inflammatory responses and play a vital role in the pathogenesis of inflammatory diseases such as allergy, asthma, and atherosclerosis. We have reported that cigarette smoke extract (CSE) significantly increased IL-6 and IL-8 production in IL-1ß-activated human mast cell line (HMC-1). Baicalein (BAI) has anti-inflammatory properties and inhibits IL-1ß- and TNF-α-induced inflammatory cytokine production from HMC-1. The goal of the present study was to examine the effect of BAI on IL-6 and IL-8 production from CSE-treated and IL-1ß-activated HMC-1. METHODS: Main-stream (Ms) and Side-stream (Ss) cigarette smoke were collected onto fiber filters and extracted in RPMI-1640 medium. Two ml of HMC-1 at 1 × 106 cells/mL were cultured with CSE in the presence or absence of IL-1ß (10 ng/mL) for 24 hrs. A group of HMC-1 cells stimulated with both IL-1ß (10 ng/ml) and CSE was also treated with BAI. The expression of IL-6 and IL-8 was assessed by ELISA and RT-PCR. NF-κB activation was measured by electrophoretic mobility shift assay (EMSA) and IκBα degradation by Western blot. RESULTS: Both Ms and Ss CSE significantly increased IL-6 and IL-8 production (p < 0.001) in IL-1ß-activated HMC-1. CSE increased NF-κB activation and decreased cytoplasmic IκBα proteins in IL-1ß-activated HMC-1. BAI (1.8 to 30 µM) significantly inhibited production of IL-6 and IL-8 in a dose-dependent manner in IL-1ß-activated HMC-1 with the optimal inhibition concentration at 30 µM, which also significantly inhibited the enhancing effect of CSE on IL-6 and IL-8 production in IL-1ß-activated HMC-1. BAI inhibited NF-κB activation and increased cytoplasmic IκBα proteins in CSE-treated and IL-1ß-activated HMC-1. CONCLUSIONS: Our results showed that CSE significantly increased inflammatory cytokines IL-6 and IL-8 production in IL-1ß-activated HMC-1. It may partially explain why cigarette smoke contributes to lung and cardiovascular diseases. BAI inhibited the production of inflammatory cytokines through inhibition of NF-κB activation and IκBα phosphorylation and degradation. This inhibitory effect of BAI on the expression of inflammatory cytokines induced by CSE suggests its usefulness in the development of novel anti-inflammatory therapies.

SELECTION OF CITATIONS
SEARCH DETAIL