Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Mol Med ; 24(1): 37, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30134825

ABSTRACT

BACKGROUND: Histone deacetylases (HDACs) play a critical role in modulating myocardial protection and cardiomyocyte survivals. However, Specific HDAC isoforms in mediating myocardial ischemia/reperfusion injury remain currently unknown. We used cardiomyocyte-specific overexpression of active HDAC4 to determine the functional role of activated HDAC4 in regulating myocardial ischemia and reperfusion in isovolumetric perfused hearts. METHODS: In this study, we created myocyte-specific active HDAC4 transgenic mice to examine the functional role of active HDAC4 in mediating myocardial I/R injury. Ventricular function was determined in the isovolumetric heart, and infarct size was determined using tetrazolium chloride staining. RESULTS: Myocyte-specific overexpressing activated HDAC4 in mice promoted myocardial I/R injury, as indicated by the increases in infarct size and reduction of ventricular functional recovery following I/R injury. Notably, active HDAC4 overexpression led to an increase in LC-3 and active caspase 3 and decrease in SOD-1 in myocardium. Delivery of chemical HDAC inhibitor attenuated the detrimental effects of active HDAC4 on I/R injury, revealing the pivotal role of active HDAC4 in response to myocardial I/R injury. CONCLUSIONS: Taken together, these findings are the first to define that activated HDAC4 as a crucial regulator for myocardial ischemia and reperfusion injury.


Subject(s)
Histone Deacetylases/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Animals , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Male , Mice, Transgenic , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/physiology , Swine , Ventricular Function, Left
2.
Cell Death Differ ; 25(5): 966-982, 2018 05.
Article in English | MEDLINE | ID: mdl-29358670

ABSTRACT

The present study investigated whether TLR3 is required for neonatal heart repair and regeneration following myocardial infarction (MI). TLR3 deficient neonatal mice exhibited impaired cardiac functional recovery and a larger infarct size, while wild type neonatal mice showed cardiac functional recovery and small infarct size after MI. The data suggest that TLR3 is essential for the regeneration and repair of damaged neonatal myocardium. In vitro treatment of neonatal cardiomyocytes with a TLR3 ligand, Poly (I:C), significantly enhances glycolytic metabolism, YAP1 activation and proliferation of cardiomyocytes which were prevented by a glycolysis inhibitor, 2-deoxyglucose (2-DG). Administration of 2-DG to neonatal mice abolished cardiac functional recovery and YAP activation after MI, suggesting that TLR3-mediated regeneration and repair of the damaged neonatal myocardium is through glycolytic-dependent YAP1 activation. Inhibition of YAP1 activation abolished Poly (I:C) induced proliferation of neonatal cardiomyocytes. Interestingly, activation of YAP1 increases the expression of miR-152 which represses the expression of cell cycle inhibitory proteins, P27kip1 and DNMT1, leading to cardiomyocyte proliferation. We conclude that TLR3 is required for neonatal heart regeneration and repair after MI. The mechanisms involve glycolytic-dependent YAP1 activation, resulting in miR-152 expression which targets DNMT1/p27kip1.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation , Glycolysis , MicroRNAs/biosynthesis , Myocardial Infarction/metabolism , Myocardium/metabolism , Phosphoproteins/metabolism , Regeneration , Toll-Like Receptor 3/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Newborn , Cell Cycle Proteins , Mice , Mice, Knockout , MicroRNAs/genetics , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Phosphoproteins/genetics , Toll-Like Receptor 3/genetics , YAP-Signaling Proteins
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.
Oncotarget ; 7(52): 86926-86936, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27894079

ABSTRACT

BACKGROUND: Myocardial apoptosis plays an important role in myocardial ischemia/reperfusion (I/R) injury. Activation of PI3K/Akt signaling protects the myocardium from I/R injury. This study investigated the role of miR-214 in hypoxia/reoxygenation (H/R)-induced cell damage in vitro and myocardial I/R injury in vivo. METHODS AND RESULTS: H9C2 cardiomyoblasts were transfected with lentivirus expressing miR-214 (LmiR-214) or lentivirus expressing scrambled miR-control (LmiR-control) respectively, to establish cell lines of LmiR-214 and LmiR-control. The cells were subjected to hypoxia for 4 h followed by reoxygenation for 24 h. Transfection of LmiR-214 suppresses PTEN expression, significantly increases the levels of Akt phosphorylation, markedly attenuates LDH release, and enhances the viability of the cells subjected to H/R. In vivo transfection of mouse hearts with LmiR-214 significantly attenuates I/R induced cardiac dysfunction and reduces I/R-induced myocardial infarct size. LmiR-214 transfection significantly attenuates I/R-induced myocardial apoptosis and caspase-3/7 and caspase-8 activity. Increased expression of miR-214 by transfection of LmiR-214 suppresses PTEN expression, increases the levels of phosphorylated Akt, represses Bim1 expression and induces Bad phosphorylation in the myocardium. In addition, in vitro data shows transfection of miR-214 mimics to H9C2 cells suppresses the expression and translocation of Bim1 from cytosol to mitochondria and induces Bad phosphorylation. CONCLUSIONS: Our in vitro and in vivo data suggests that miR-214 protects cells from H/R induced damage and attenuates I/R induced myocardial injury. The mechanisms involve activation of PI3K/Akt signaling by targeting PTEN expression, induction of Bad phosphorylation, and suppression of Bim1 expression, resulting in decreases in I/R-induced myocardial apoptosis.


Subject(s)
Bcl-2-Like Protein 11/genetics , MicroRNAs/genetics , Myocardial Reperfusion Injury/genetics , Myocytes, Cardiac/metabolism , PTEN Phosphohydrolase/genetics , Animals , Apoptosis/genetics , Bcl-2-Like Protein 11/metabolism , Caspases/metabolism , Cell Hypoxia , Cell Line , Gene Expression Regulation , Male , Mice, Inbred C57BL , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/cytology , Oxygen/metabolism , PTEN Phosphohydrolase/metabolism , Phosphorylation , Rats , Signal Transduction/genetics , bcl-Associated Death Protein/metabolism
5.
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
6.
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
7.
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
8.
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
9.
Shock ; 42(1): 31-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24569510

ABSTRACT

Cardiac dysfunction is a major consequence that contributes to the high mortality of trauma-hemorrhage (TH) patients. Recent evidence suggests that innate immune and inflammatory responses mediated by Toll-like receptors (TLRs) play a critical role in the pathophysiologic mechanisms of acute organ dysfunction during TH. This study investigated the role of TLR4 in cardiac dysfunction following TH. Toll-like receptor 4-deficient (TLR4-/-, n = 7/group) and age-matched wild-type (WT, n = 8/group) mice were subjected to TH that was induced by soft tissue injury and blood withdrawal from the jugular vein to a mean arterial pressure of 35 ± 5 mmHg. Cardiac function and mean arterial pressure were measured with a Millar system before, during, and after blood withdrawal. Sham surgical-operated mice served as control (WT, n = 9/group; TLR4-/-, n = 10/group). Cardiac function in WT mice was significantly reduced following TH. However, cardiac function was well preserved in TLR4-/- mice. Administration of a TLR4 antagonist (3 mg/kg) to WT mice also significantly attenuated TH-induced cardiac dysfunction. Western blot showed that either TLR4-/- or TLR4 antagonist markedly attenuated TH-induced decreases in the levels of phosphorylated-Akt in myocardium. In addition, inhibition of TLR4 attenuated TH-induced myocardial nuclear factor κB-binding activity as well as lung myeloperoxidase activity and tumor necrosis factor α production. The data indicate that TLR4 plays a central role in TH-induced cardiac dysfunction. Toll-like receptor 4 deficiency or TLR4 inhibition attenuated cardiac dysfunction following TH, which may involve activation of the phosphoinositide 3-kinase/Akt signaling and decrease in nuclear factor κB-binding activity. Toll-like receptor 4 antagonism may be a new and novel approach for the treatment and management of cardiac dysfunction in TH patients.


Subject(s)
Heart/physiopathology , Shock, Hemorrhagic/physiopathology , Toll-Like Receptor 4/physiology , Animals , Dose-Response Relationship, Drug , Hemodynamics/drug effects , Hemodynamics/physiology , Lipid A/analogs & derivatives , Lipid A/pharmacology , Male , Mice, Knockout , NF-kappa B/metabolism , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/physiology , Phosphorylation/drug effects , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/metabolism , Shock, Hemorrhagic/etiology , Shock, Hemorrhagic/metabolism , Soft Tissue Injuries/complications , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/deficiency , Tumor Necrosis Factor-alpha/biosynthesis
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.
Microsc Microanal ; 19(6): 1428-35, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24029590

ABSTRACT

The urinary bladder is a unique organ in that its normal function is storage and release of urine, and vasculature in its wall exhibits specialized features designed to accommodate changes in pressure with emptying and filling. Although we have previously described the fine details of the microvasculature of the urinary bladder of the rabbit and dog, information on the fine details of the microvasculature of the mouse bladder were deemed to be of value because of the increasing use of this species in developing genetic models for studying human disorders. The present study shows that many of the special features of the microvasculature of the mouse urinary bladder are similar to those described in the rabbit and dog, including vessel coiling, abundant collateral circulation, arterial sphincters, and a dense mucosal capillary plexus.


Subject(s)
Microvessels/anatomy & histology , Urinary Bladder/anatomy & histology , Animals , Corrosion Casting , Mice , Microscopy
12.
Methods Mol Biol ; 1036: 1-17, 2013.
Article in English | MEDLINE | ID: mdl-23807782

ABSTRACT

Cellular cardiomyoplasty is a cell therapy using stem cells or progenitor cells for myocardial regeneration to improve cardiac function and mitigate heart failure. Since we first published cellular cardiomyoplasty in 1989, this procedure became the innovative method to treat damaged myocardium other than heart transplantation. A significant improvement in cardiac function, metabolism, and perfusion is generally observed in experimental and clinical studies, but the improvement is mild and incomplete. Although safety, feasibility, and efficacy have been well documented for the procedure, the beneficial mechanisms remain unclear and optimization of the procedure requires further study. This chapter briefly reviews the stem cells used for cellular cardiomyoplasty and their clinical outcomes with possible improvements in future studies.


Subject(s)
Cardiomyoplasty/methods , Heart Diseases/therapy , Stem Cell Transplantation , Stem Cells/cytology , Humans
13.
Methods Mol Biol ; 1036: 19-32, 2013.
Article in English | MEDLINE | ID: mdl-23807783

ABSTRACT

Skeletal muscle satellite cells (myoblasts) are the primary stem cells of skeletal muscle which contribute to growth, maintenance, and repair of the muscles. Satellite cells are the first stem cells used for cellular cardiomyoplasty more than 20 years ago. The isolation, culture, labeling, and identification of satellite cells are described in detail here. The implantation and outcomes of cellular cardiomyoplasty using satellite cells have been summarized in the previous chapter (Chapter 1).


Subject(s)
Cardiomyoplasty/methods , Cell Separation/methods , Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Animals , Cell Differentiation , Cells, Cultured , Humans , In Situ Hybridization, Fluorescence , Muscle, Skeletal/physiology , Regeneration , Satellite Cells, Skeletal Muscle/physiology
14.
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
15.
PLoS One ; 7(9): e44712, 2012.
Article in English | MEDLINE | ID: mdl-23028587

ABSTRACT

Phosphoinositide-3-kinase (PI3K)/Akt dependent signaling has been shown to improve outcome in sepsis/septic shock. There is also ample evidence that PI3K/Akt dependent signaling plays a crucial role in maintaining normal cardiac function. We hypothesized that PI3K/Akt signaling may ameliorate septic shock by attenuating sepsis-induced cardiac dysfunction. Cardiac function and survival were evaluated in transgenic mice with cardiac myocyte specific expression of constitutively active PI3K isoform, p110α (caPI3K Tg). caPI3K Tg and wild type (WT) mice were subjected to cecal ligation/puncture (CLP) induced sepsis. Wild type CLP mice showed dramatic cardiac dysfunction at 6 hrs. Septic cardiomyopathy was significantly attenuated in caPI3K CLP mice. The time to 100% mortality was 46 hrs in WT CLP mice. In contrast, 80% of the caPI3K mice survived at 46 hrs after CLP (p<0.01) and 50% survived >30 days (p<0.01). Cardiac caPI3K expression prevented expression of an inflammatory phenotype in CLP sepsis. Organ neutrophil infiltration and lung apoptosis were also effectively inhibited by cardiac PI3k p110α expression. Cardiac high mobility group box-1 (HMGB-1) translocation was also inhibited by caPI3K p110α expression. We conclude that cardiac specific activation of PI3k/Akt dependent signaling can significantly modify the morbidity and mortality associated with sepsis. Our data also indicate that myocardial function/dysfunction plays a prominent role in the pathogenesis of sepsis and that maintenance of cardiac function during sepsis is essential. Finally, these data suggest that modulation of the PI3K/p110α signaling pathway may be beneficial in the prevention and/or management of septic cardiomyopathy and septic shock.


Subject(s)
Myocardium/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Sepsis/enzymology , Sepsis/therapy , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cytokines/blood , Echocardiography , Hemodynamics/physiology , Mice , Mice, Transgenic , Peroxidase/metabolism , Phosphatidylinositol 3-Kinase/genetics , Sepsis/microbiology
16.
Shock ; 38(2): 146-52, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22576005

ABSTRACT

Cardiovascular collapse is the major factor contributing to the mortality of trauma-hemorrhage (T-H) patients. Toll-like receptors (TLRs) play a critical role in T-H-induced cardiac dysfunction. This study evaluated the role of TLR9 agonist, CpG-oligodeoxynucleotide (ODN) 1826, in cardiac functional recovery after T-H. Trauma-hemorrhage was induced in a murine model by soft tissue injury and blood withdrawals from the jugular vein to a mean arterial pressure of 35 ± 5 mmHg. Mice were treated with CpG-ODN 1826 (10 µg/30 g body weight) by intraperitoneal injection 1 h before T-H (n = 5-8/group). Hemodynamic parameters were measured before, during hemorrhage, and at 60 min after T-H. Trauma-hemorrhage significantly decreased the mean arterial pressure and left ventricular pressure compared with sham controls. In contrast, CpG-ODN administration significantly attenuated the decrease in arterial pressure and left ventricular pressure due to T-H. Trauma-hemorrhage markedly decreased myocardial levels of phosphorylated Akt by 57.9%. However, CpG-ODN treatment significantly blunted the decrement in phospho-Akt by activating the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. The PI3K inhibitor LY294002 partially abolished CpG-induced cardioprotection, indicating that additional signaling pathways are involved in the protective effect of CpG-ODN after T-H. We observed that CpG-ODN treatment also significantly attenuated the decrease in myocardial phospho-ERK levels after T-H. Inhibition of ERK by U0126 also partially abolished the cardioprotective effect of CpG-ODN after T-H. Our data suggest that CpG-ODN significantly attenuates T-H-induced cardiac dysfunction. The mechanisms involve activation of both PI3K/Akt and ERK signaling pathways. The TLR9 agonist, CpG-ODN 1826, may provide a novel treatment strategy for preventing or managing cardiac dysfunction and enhancing recovery in T-H patients.


Subject(s)
Adjuvants, Immunologic/pharmacology , Heart Diseases/prevention & control , Hemorrhage/complications , Hypotension/prevention & control , Oligodeoxyribonucleotides/pharmacology , Soft Tissue Injuries/complications , Toll-Like Receptor 9/agonists , Animals , Chromones/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism
17.
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
18.
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
19.
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
20.
Am J Physiol Heart Circ Physiol ; 301(3): H848-55, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21642503

ABSTRACT

Myocardial dysfunction is a major consequence of septic shock and contributes to the high mortality of sepsis. High-mobility group box 1 (HMGB1) serves as a late mediator of lethality in sepsis. We have reported that glucan phosphate (GP) attenuates cardiac dysfunction and increases survival in cecal ligation and puncture (CLP)-induced septic mice. In the present study, we examined the effect of GP on HMGB1 translocation from the nucleus to the cytoplasm in the myocardium of septic mice. GP was administered to mice 1 h before induction of CLP. Sham-operated mice served as control. The levels of HMGB1, Toll-like receptor 4 (TLR4), and NF-κB binding activity were examined. In an in vitro study, H9C2 cardiomyoblasts were treated with lipopolysaccharide (LPS) in the presence or absence of GP. H9C2 cells were also transfected with Ad5-IκBα mutant, a super repressor of NF-κB activity, before LPS stimulation. CLP significantly increased the levels of HMGB1, TLR4, and NF-κB binding activity in the myocardium. In contrast, GP administration attenuated CLP-induced HMGB1 translocation from the nucleus to the cytoplasm and reduced CLP-induced increases in TLR4 and NF-κB activity in the myocardium. In vitro studies showed that GP prevented LPS-induced HMGB1 translocation and NF-κB binding activity. Blocking NF-κB binding activity by Ad5-IκBα attenuated LPS-induced HMGB1 translocation. GP administration also reduced the LPS-stimulated interaction of HMGB1 with TLR4. These data suggest that attenuation of HMGB1 translocation by GP is mediated through inhibition of NF-κB activation in CLP-induced sepsis and that activation of NF-κB is required for HMGB1 translocation.


Subject(s)
Glucans/pharmacology , HMGB1 Protein/metabolism , Myocytes, Cardiac/drug effects , NF-kappa B/metabolism , Sepsis/prevention & control , Analysis of Variance , Animals , Cecum/microbiology , Cecum/surgery , Cell Line , Disease Models, Animal , I-kappa B Proteins/metabolism , Ligation , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Mutation , Myocytes, Cardiac/metabolism , NF-KappaB Inhibitor alpha , Protein Transport , Punctures , Rats , Sepsis/genetics , Sepsis/metabolism , Sepsis/microbiology , Severity of Illness Index , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...