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










Publication year range
1.
Life Sci ; 332: 122113, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37739163

ABSTRACT

AIMS: Ferroptosis has grown in importance as a key factor in ischemia-reperfusion (I/R) injury. This study explores the mechanism underlying fibrotic scarring extending along myofibers in cardiac ischemic injury and demonstrates the integral role of ferroptosis in causing a unique cell death pattern linked to I/R injury. MAIN METHODS: Cadaveric hearts from individuals who had ischemic injury were examined by histological assays. We created a novel model of inducing cell death in H9c2 cells, and used it to demonstrate ferroptotic cell death extending in a cell-to-cell manner. Ex vivo Langendorff-perfused hearts were used alongside the model to replicate cell death extension along myofibers while also demonstrating protective effects of a ferroptosis inhibitor, ferrostatin-1 (Fer-1). KEY FINDINGS: Human hearts from individuals who had I/R injury demonstrated scarring along myofibers that was consistent with mouse models, suggesting that cell death extended from cell-to-cell. Treatment with Ras-selective lethal 3 (RSL3), a ferroptosis inducer, and exposure to excess iron exacerbated cell death propagation in in vitro models, and inhibition of ferroptosis by Fer-1 blunted this effect in both settings. In ex vivo models, Fer-1 was sufficient to reduce cell death along the myofibers caused by external injury. SIGNIFICANCE: The unique I/R injury-induced pattern of cell death along myofibers requires novel injury models that mimic this phenomenon, thus we established new methods to replicate it. Ferroptosis is important in propagating injury between cells and better understanding this mechanism may lead to therapeutic responses that limit I/R injury.


Subject(s)
Ferroptosis , Heart Injuries , Reperfusion Injury , Animals , Mice , Humans , Myocytes, Cardiac , Cicatrix , Cell Death
2.
Physiol Rep ; 9(6): e14807, 2021 03.
Article in English | MEDLINE | ID: mdl-33769701

ABSTRACT

The mechanistic target of rapamycin (mTOR) is a key mediator of energy metabolism, cell growth, and survival. While previous studies using transgenic mice with cardiac-specific overexpression of mTOR (mTOR-Tg) demonstrated the protective effects of cardiac mTOR against ischemia-reperfusion (I/R) injury in both ex vivo and in vivo models, the mechanisms underlying the role of cardiac mTOR in cardiac function following I/R injury are not well-understood. Torin1, a pharmacological inhibitor of mTOR complex (mTORC) 1 and mTORC2, significantly decreased functional recovery of LV developed pressure in ex vivo I/R models (p < 0.05). To confirm the role of mTOR complexes in I/R injury, we generated cardiac-specific mTOR-knockout (CKO) mice. In contrast to the effects of Torin1, CKO hearts recovered better after I/R injury than control hearts (p < 0.05). Interestingly, the CKO hearts had exhibited irregular contractions during the reperfusion phase. Calcium is a major factor in Excitation-Contraction (EC) coupling via Sarcoplasmic Reticulum (SR) calcium release. Calcium is also key in opening the mitochondrial permeability transition pore (mPTP) and cell death following I/R injury. Caffeine-induced SR calcium release in isolated CMs showed that total SR calcium content was lower in CKO than in control CMs. Western blotting showed that a significant amount of mTOR localizes to the SR/mitochondria and that GSK3-ß phosphorylation, a key factor in SR calcium mobilization, was decreased. These findings suggest that cardiac mTOR located to the SR/mitochondria plays a vital role in EC coupling and cell survival in I/R injury.


Subject(s)
Calcium Signaling , Heart/physiopathology , Myocardial Reperfusion Injury/physiopathology , TOR Serine-Threonine Kinases/physiology , Animals , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/physiology , Sarcoplasmic Reticulum/physiology , TOR Serine-Threonine Kinases/genetics
3.
Life Sci ; 232: 116665, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31323273

ABSTRACT

AIMS: Overexpression of the mechanistic target of rapamycin (mTOR), a member of the PIKK (phosphoinositide kinase-related kinase) family, protects cardiomyocytes from cell death induced by pathological stimuli such as ischemia. We previously reported that posttranslational modification of mTOR plays an important role in regulating cardiac mTOR expression. The aim of this study was to see if Tel2 (telomere maintenance 2), a protein that regulates the abundance of PIKKs, confers similar cardioprotective effects as mTOR. Tel2 is not well-characterized in cardiomyocytes, therefore we examined the effects of Tel2 on cardiomyocyte viability under ischemic stress conditions. MATERIALS AND METHODS: We overexpressed Tel2 or silenced Tel2 with siRNA in the HL-1 cardiomyocyte cell line to survey the effects of Tel2 overexpression and downregulation on cell survival during hypoxia. Adult mouse cardiomyocytes transfected with Tel2 adenoviruses were used to test whether Tel2 sufficiently prevented cardiomyocyte cell death against hydrogen peroxide (H2O2). KEY FINDINGS: Overexpressing Tel2 increased mTOR expression with a concomitant increase in mTOR Complex 1 (mTORC1) and mTORC2 activity in HL-1 cells. Tel2 deletion decreased mTOR expression, and mTORC1 and mTORC2 activity accordingly. In both HL-1 cells and adult mouse cardiomyocytes, Tel2 overexpression protected cardiomyocytes under ischemic stress. These effects were mTOR-dependent, as mTOR inhibitors blunted the effects of Tel2. While gene silencing of Tel2 did not affect cell survival under normoxia, Tel2 silencing made cardiomyocytes more vulnerable to cell death under hypoxia. SIGNIFICANCE: Upregulating Tel2 expression increases mTOR-mediated cardiomyocyte survival and targeting Tel2 could be another therapeutic strategy against ischemic heart disease.


Subject(s)
Cell Survival/physiology , Myocytes, Cardiac/cytology , Telomere-Binding Proteins/physiology , Adenoviridae/genetics , Animals , Cell Death/drug effects , Cell Line , Gene Silencing , Hydrogen Peroxide/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Signal Transduction , Telomere-Binding Proteins/genetics , Transfection
5.
Curr Drug Targets ; 19(9): 1068-1076, 2018.
Article in English | MEDLINE | ID: mdl-29874997

ABSTRACT

Iron is an essential mineral required for a variety of vital biological functions. Despite being vital for life, iron also has potentially toxic aspects. Iron has been investigated as a risk factor for coronary artery disease (CAD), however, iron's toxicity in CAD patients still remains controversial. One possible mechanism behind the toxicity of iron is "ferroptosis", a newly described form of irondependent cell death. Ferroptosis is an iron-dependent form of regulated cell death that is distinct from apoptosis, necroptosis, and other types of cell death. Ferroptosis has been reported in ischemiareperfusion (I/R) injury and several other diseases. Recently, we reported that ferroptosis is a significant form of cell death in cardiomyocytes. Moreover, myocardial hemorrhage, a major event in the pathogenesis of heart failure, could trigger the release of free iron into cardiac muscle and is an independent predictor of adverse left ventricular remodeling after myocardial infarction. Iron deposition in the heart can now be detected with advanced imaging methods, such as T2 star (T2*) cardiac magnetic resonance imaging, which can non-invasively predict iron levels in the myocardium and detect myocardial hemorrhage, thus existing technology could be used to assess myocardial iron. We will discuss the role of iron in cardiovascular diseases and especially with regard to myocardial I/R injury.


Subject(s)
Cardiovascular Diseases/physiopathology , Coronary Artery Disease/physiopathology , Iron/toxicity , Animals , Cell Death/physiology , Heart Failure/physiopathology , Hemorrhage/physiopathology , Humans , Iron/analysis , Iron/metabolism , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/pathology , Risk Factors , Ventricular Remodeling/physiology
6.
Sci Rep ; 8(1): 7601, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29765083

ABSTRACT

Kawasaki disease (KD), which is the leading cause of pediatric heart disease, is characterized by coronary vasculitis and subsequent aneurysm formation. Although intravenous immunoglobulin therapy is effective for reducing aneurysm formation, a certain number of patients are resistant to this therapy. Because interleukin-10 (IL-10) was identified as a negative regulator of cardiac inflammation in a murine model of KD induced by Candida albicans water-soluble fraction (CAWS), we investigated the effect of IL-10 supplementation in CAWS-induced vasculitis. Mice were injected intramuscularly with adeno-associated virus (AAV) vector encoding IL-10, then treated with CAWS. The induction of AAV-mediated IL-10 (AAV-IL-10) significantly attenuated the vascular inflammation and fibrosis in the aortic root and coronary artery, resulting in the improvement of cardiac dysfunction and lethality. The predominant infiltrating inflammatory cells in the vascular walls were Dectin-2+CD11b+ macrophages. In vitro experiments revealed that granulocyte/macrophage colony-stimulating factor (GM-CSF) induced Dectin-2 expression in bone marrow-derived macrophages and enhanced the CAWS-induced production of tumor necrosis factor-α (TNF-α) and IL-6. IL-10 had no effect on the Dectin-2 expression but significantly inhibited the production of cytokines. IL-10 also inhibited CAWS-induced phosphorylation of ERK1/2, but not Syk. Furthermore, the induction of AAV-IL-10 prevented the expression of TNF-α and IL-6, but not GM-CSF and Dectin-2 at the early phase of CAWS-induced vasculitis. These findings demonstrate that AAV-IL-10 may have therapeutic application in the prevention of coronary vasculitis and aneurysm formation, and provide new insights into the mechanism underlying the pathogenesis of KD.


Subject(s)
Candida albicans/chemistry , Genetic Vectors/administration & dosage , Interleukin-10/genetics , Mucocutaneous Lymph Node Syndrome/therapy , Vasculitis/therapy , Animals , Dependovirus/genetics , Disease Models, Animal , Heart Function Tests/drug effects , Humans , Injections, Intramuscular , Macrophages/metabolism , Mice , Mucocutaneous Lymph Node Syndrome/etiology , Mucocutaneous Lymph Node Syndrome/genetics , Mucocutaneous Lymph Node Syndrome/physiopathology , Treatment Outcome , Vasculitis/etiology , Vasculitis/physiopathology
7.
Am J Physiol Heart Circ Physiol ; 314(3): H659-H668, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29127238

ABSTRACT

Clinical studies have suggested that myocardial iron is a risk factor for left ventricular remodeling in patients after myocardial infarction. Ferroptosis has recently been reported as a mechanism of iron-dependent nonapoptotic cell death. However, ferroptosis in the heart is not well understood. Mechanistic target of rapamycin (mTOR) protects the heart against pathological stimuli such as ischemia. To define the role of cardiac mTOR on cell survival in iron-mediated cell death, we examined cardiomyocyte (CM) cell viability under excess iron and ferroptosis conditions. Adult mouse CMs were isolated from cardiac-specific mTOR transgenic mice, cardiac-specific mTOR knockout mice, or control mice. CMs were treated with ferric iron [Fe(III)]-citrate, erastin, a class 1 ferroptosis inducer, or Ras-selective lethal 3 (RSL3), a class 2 ferroptosis inducer. Live/dead cell viability assays revealed that Fe(III)-citrate, erastin, and RSL3 induced cell death. Cotreatment with ferrostatin-1, a ferroptosis inhibitor, inhibited cell death in all conditions. mTOR overexpression suppressed Fe(III)-citrate, erastin, and RSL3-induced cell death, whereas mTOR deletion exaggerated cell death in these conditions. 2',7'-Dichlorodihydrofluorescein diacetate measurement of reactive oxygen species (ROS) production showed that erastin-induced ROS production was significantly lower in mTOR transgenic versus control CMs. These findings suggest that ferroptosis is a significant type of cell death in CMs and that mTOR plays an important role in protecting CMs against excess iron and ferroptosis, at least in part, by regulating ROS production. Understanding the effects of mTOR in preventing iron-mediated cell death will provide a new therapy for patients with myocardial infarction. NEW & NOTEWORTHY Ferroptosis has recently been reported as a new form of iron-dependent nonapoptotic cell death. However, ferroptosis in the heart is not well characterized. Using cultured adult mouse cardiomyocytes, we demonstrated that the mechanistic target of rapamycin plays an important role in protecting cardiomyocytes against excess iron and ferroptosis.


Subject(s)
Iron/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/enzymology , TOR Serine-Threonine Kinases/metabolism , Animals , Carbolines/toxicity , Cell Death , Cell Survival , Cells, Cultured , Cyclohexylamines/toxicity , Ferric Compounds/toxicity , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phenylenediamines/toxicity , Piperazines/toxicity , Reactive Oxygen Species/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics
8.
PLoS One ; 12(5): e0176676, 2017.
Article in English | MEDLINE | ID: mdl-28493895

ABSTRACT

Cardiac glycosides such as digoxin are Na+/K+-ATPase inhibitors that are widely used for the treatment of chronic heart failure and cardiac arrhythmias; however, recent epidemiological studies have suggested a relationship between digoxin treatment and increased mortality. We previously showed that nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasomes, which regulate caspase-1-dependent interleukin (IL)-1ß release, mediate the sterile cardiovascular inflammation. Because the Na+/K+-ATPase is involved in inflammatory responses, we investigated the role of NLRP3 inflammasomes in the pathophysiology of cardiac glycoside-induced cardiac inflammation and dysfunction. The cardiac glycoside ouabain induced cardiac dysfunction and injury in wild-type mice primed with a low dose of lipopolysaccharide (LPS), although no cardiac dysfunction was observed in mice treated with either ouabain or LPS alone. Ouabain also induced cardiac inflammatory responses, such as macrophage infiltration and IL-1ß release, when mice were primed with LPS. These cardiac manifestations were all significantly attenuated in mice deficient in IL-1ß. Furthermore, deficiency of NLRP3 inflammasome components, NLRP3 and caspase-1, also attenuated ouabain-induced cardiac dysfunction and inflammation. In vitro experiments revealed that ouabain induced NLRP3 inflammasome activation as well as subsequent IL-1ß release from macrophages, and this activation was mediated by K+ efflux. Our findings demonstrate that cardiac glycosides promote cardiac inflammation and dysfunction through NLRP3 inflammasomes and provide new insights into the mechanisms underlying the adverse effects of cardiac glycosides.


Subject(s)
Inflammasomes/metabolism , Inflammation/pathology , Inflammation/physiopathology , Myocardium/metabolism , Myocardium/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Ouabain/adverse effects , Animals , Biological Transport/drug effects , Caspase 1/metabolism , Interleukin-1beta/deficiency , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Myocardium/enzymology , Potassium/metabolism
9.
Am J Physiol Endocrinol Metab ; 311(5): E881-E890, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27702746

ABSTRACT

Caspase-1 is a cysteine protease responsible for the processing of the proinflammatory cytokine interleukin-1ß and activated by the formation of inflammasome complexes. Although several investigations have found a link between diet-induced obesity and caspase-1, the relationship remains controversial. Here, we found that mice deficient in caspase-1 were susceptible to high-fat diet-induced obesity with increased adiposity as well as normal lipid and glucose metabolism. Caspase-1 deficiency clearly promoted the infiltration of inflammatory macrophages and increased the production of C-C motif chemokine ligand 2 (CCL2) in the adipose tissue. The dominant cellular source of CCL2 was stromal vascular fraction rather than adipocytes in the adipose tissue. These findings demonstrate a critical role of caspase-1 in macrophage-driven inflammation in the adipose tissue and the development of obesity. These data provide novel insights into the mechanisms underlying inflammation in the pathophysiology of obesity.


Subject(s)
Adipose Tissue/immunology , Caspase 1/genetics , Chemokine CCL2/immunology , Macrophages/immunology , Obesity/genetics , Adipocytes/immunology , Adipocytes/pathology , Adiponectin/immunology , Adipose Tissue/pathology , Animals , Blood Glucose/metabolism , Body Composition , Caspase 1/immunology , Cholesterol/metabolism , Diet, High-Fat , Flow Cytometry , Gene Expression Profiling , Glucose Tolerance Test , Insulin/metabolism , Interferon-gamma/immunology , Interleukin-12/immunology , Interleukin-18/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Leptin/immunology , Male , Mice , Mice, Knockout , Obesity/immunology , Obesity/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/immunology , X-Ray Microtomography
10.
Sci Rep ; 6: 26489, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27225830

ABSTRACT

NLRP3 inflammasomes recognize non-microbial danger signals and induce release of proinflammatory cytokine interleukin (IL)-1ß, leading to sterile inflammation in cardiovascular disease. Because sterile inflammation is involved in doxorubicin (Dox)-induced cardiotoxicity, we investigated the role of NLRP3 inflammasomes in Dox-induced cardiotoxicity. Cardiac dysfunction and injury were induced by low-dose Dox (15 mg/kg) administration in NLRP3-deficient (NLRP3(-/-)) mice but not in wild-type (WT) and IL-1ß(-/-) mice, indicating that NLRP3 deficiency enhanced the susceptibility to Dox-induced cardiotoxicity independent of IL-1ß. Although the hearts of WT and NLRP3(-/-) mice showed no significant difference in inflammatory cell infiltration, macrophages were the predominant inflammatory cells in the hearts, and cardiac IL-10 production was decreased in Dox-treated NLRP3(-/-) mice. Bone marrow transplantation experiments showed that bone marrow-derived cells contributed to the exacerbation of Dox-induced cardiotoxicity in NLRP3(-/-) mice. In vitro experiments revealed that NLRP3 deficiency decreased IL-10 production in macrophages. Furthermore, adeno-associated virus-mediated IL-10 overexpression restored the exacerbation of cardiotoxicity in the NLRP3(-/-) mice. These results demonstrated that NLRP3 regulates macrophage IL-10 production and contributes to the pathophysiology of Dox-induced cardiotoxicity, which is independent of IL-1ß. Our findings identify a novel role of NLRP3 and provided new insights into the mechanisms underlying Dox-induced cardiotoxicity.


Subject(s)
Doxorubicin/toxicity , Heart Injuries/immunology , Interleukin-10/metabolism , Interleukin-1beta/genetics , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Animals , Bone Marrow Transplantation/adverse effects , Cardiotoxicity , Cells, Cultured , Disease Models, Animal , Heart Injuries/chemically induced , Heart Injuries/genetics , Humans , Interleukin-1beta/deficiency , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
11.
Sci Rep ; 5: 15883, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26515636

ABSTRACT

Inflammation plays an important role in the development of obesity and metabolic disorders; however, it has not been fully understood how inflammation occurs and is regulated in their pathogenesis. Low-molecular mass protein-7 (LMP7) is a proteolytic subunit of the immunoproteasome that shapes the repertoire of antigenic peptides on major histocompatibility complex class I molecule. In this study, we investigated the role of LMP7 in the development of obesity and metabolic disorders using LMP7-deficient mice. LMP7 deficiency conveyed resistant to obesity, and improved glucose intolerance and insulin sensitivity in mice fed with high-fat diet (HFD). LMP7 deficiency decreased pancreatic lipase expression, increased fecal lipid contents, and inhibited the increase of plasma triglyceride levels upon oral oil administration or HFD feeding. Using bone marrow-transferred chimeric mice, we found that LMP7 in both bone marrow- and non-bone marrow-derived cells contributes to the development of HFD-induced obesity. LMP7 deficiency decreased inflammatory responses such as macrophage infiltration and chemokine expression while it increased serum adiponection levels. These findings demonstrate a novel role for LMP7 and provide new insights into the mechanisms underlying inflammation in the pathophysiology of obesity and metabolic disorders.


Subject(s)
Metabolic Diseases/pathology , Obesity/pathology , Proteasome Endopeptidase Complex/genetics , Adiponectin/blood , Adipose Tissue/metabolism , Animals , Antigens, CD/metabolism , Bone Marrow Transplantation , Chemokines/metabolism , Diet, High-Fat , Energy Metabolism , Insulin Resistance , Lipase/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Obesity/metabolism , Pancreas/enzymology , Proteasome Endopeptidase Complex/deficiency , Subcutaneous Fat, Abdominal/diagnostic imaging , Triglycerides/blood , X-Ray Microtomography
12.
Endocrinology ; 156(11): 4281-92, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26360504

ABSTRACT

Preeclampsia is a pregnancy-specific syndrome characterized by elevated blood pressure, proteinuria, and intrauterine growth restriction (IUGR). Although sterile inflammation appears to be involved, its pathogenesis remains unclear. Recent evidence indicates that sterile inflammation is mediated through the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasomes, composed of NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and caspase-1. Here we investigated the role of the NLRP3 inflammasomes in the pathogenesis of preeclampsia using Nlrp3(-/-) and Asc(-/-) (Nlrp3 and Asc deficient) pregnant mice. During pregnancy in mice, continuous infusion of high-dose angiotensin II (AngII) induced hypertension, proteinuria, and IUGR, whereas infusion of low-dose AngII caused hypertension alone. AngII-induced hypertension was prevented in Nlrp3(-/-) mice but not in Asc(-/-), indicating that NLRP3 contributes to gestational hypertension independently of ASC-mediated inflammasomes. Although NLRP3 deficiency had no effect on IUGR, it restored the IL-6 up-regulation in the placenta and kidney of AngII-infused mice. Furthermore, treatment with hydralazine prevented the development of gestational hypertension but not IUGR or IL-6 expression in the placenta and kidney. These findings demonstrate that NLRP3 contributes to the development of gestational hypertension independently of the inflammasomes and that IUGR and kidney injury can occur independent of blood pressure elevation during pregnancy.


Subject(s)
Blood Pressure/physiology , Carrier Proteins/metabolism , Fetal Growth Retardation/metabolism , Hypertension/metabolism , Angiotensin II , Animals , Blood Pressure/drug effects , Carrier Proteins/genetics , Disease Models, Animal , Female , Fetal Growth Retardation/genetics , Hypertension/chemically induced , Hypertension/genetics , Hypertension, Pregnancy-Induced/chemically induced , Hypertension, Pregnancy-Induced/genetics , Hypertension, Pregnancy-Induced/metabolism , Inflammasomes/metabolism , Interleukin-6/metabolism , Kidney/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Placenta/metabolism , Pregnancy
13.
Sci Rep ; 5: 10901, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-26045078

ABSTRACT

Rhabdomyolysis is one of the main causes of community-acquired acute kidney injury (AKI). Although inflammation is involved in the pathogenesis of rhabdomyolysis-induced AKI (RIAKI), little is known about the mechanism that triggers inflammation during RIAKI. Recent evidence has indicated that sterile inflammation triggered by tissue injury can be mediated through multiprotein complexes called the inflammasomes. Therefore, we investigated the role of NLRP3 inflammasomes in the pathogenesis of RIAKI using a glycerol-induced murine rhabdomyolysis model. Inflammasome-related molecules were upregulated in the kidney of RIAKI. Renal tubular injury and dysfunction preceded leukocyte infiltration into the kidney during the early phase of RIAKI, and they were markedly attenuated in mice deficient in NLRP3, ASC, caspase-1, and interleukin (IL)-1ß compared with those in wild-type mice. No difference in leukocyte infiltration was observed between wild-type and NLRP3-deficient mice. Furthermore, NLRP3 deficiency strikingly suppressed the expression of renal injury markers and inflammatory cytokines and apoptosis of renal tubular cells. These results demonstrated that NLRP3 inflammasomes contribute to inflammation, apoptosis, and tissue injury during the early phase of RIAKI and provide new insights into the mechanism underlying the pathogenesis of RIAKI.


Subject(s)
Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Carrier Proteins/metabolism , Inflammasomes/metabolism , Rhabdomyolysis/complications , Acute Kidney Injury/pathology , Animals , Apoptosis , Biomarkers , Carrier Proteins/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Inflammation Mediators/metabolism , Kidney Tubules/metabolism , Leukocytes/metabolism , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein
14.
FEBS Open Bio ; 5: 348-56, 2015.
Article in English | MEDLINE | ID: mdl-25973362

ABSTRACT

Increasing evidence indicates that caspase recruitment domain (CARD)-mediated caspase-1 (CASP1) assembly is an essential process for its activation and subsequent interleukin (IL)-1ß release, leading to the initiation of inflammation. Both CARD16 and CARD17 were previously reported as inhibitory homologs of CASP1; however, their molecular function remains unclear. Here, we identified that oligomerization activity allows CARD16 to function as a CASP1 activator. We investigated the molecular characteristics of CARD16 and CARD17 in transiently transfected HeLa cells. Although both CARD16 and CARD17 interacted with CASP1CARD, only CARD16 formed a homo-oligomer. Oligomerized CARD16 formed a filament-like structure with CASP1CARD and a speck with apoptosis-associated speck-like protein containing a CARD. A filament-like structure formed by CARD16 promoted CASP1 filament assembly and IL-1ß release. In contrast, CARD17 did not form a homo-oligomer or filaments and inhibited CASP1-dependent IL-1ß release. Mutated CARD16D27G, mimicking the CARD17 amino acid sequence, formed a homo-oligomer but failed to form a filament-like structure. Consequently, CARD16D27G weakly promoted CASP1 filament assembly and subsequent IL-1ß release. These results suggest that oligomerized CARD16 promotes CARD-mediated molecular assembly and CASP1 activation.

16.
J Biol Chem ; 290(8): 5065-5077, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25548278

ABSTRACT

Supplemental oxygen inhalation is frequently used to treat severe respiratory failure; however, prolonged exposure to hyperoxia causes hyperoxic acute lung injury (HALI), which induces acute respiratory distress syndrome and leads to high mortality rates. Recent investigations suggest the possible role of NLRP3 inflammasomes, which regulate IL-1ß production and lead to inflammatory responses, in the pathophysiology of HALI; however, their role is not fully understood. In this study, we investigated the role of NLRP3 inflammasomes in mice with HALI. Under hyperoxic conditions, NLRP3(-/-) mice died at a higher rate compared with wild-type and IL-1ß(-/-) mice, and there was no difference in IL-1ß production in their lungs. Under hyperoxic conditions, the lungs of NLRP3(-/-) mice exhibited reduced inflammatory responses, such as inflammatory cell infiltration and cytokine expression, as well as increased and decreased expression of MMP-9 and Bcl-2, respectively. NLRP3(-/-) mice exhibited diminished expression and activation of Stat3, which regulates MMP-9 and Bcl-2, in addition to increased numbers of apoptotic alveolar epithelial cells. In vitro experiments revealed that alveolar macrophages and neutrophils promoted Stat3 activation in alveolar epithelial cells. Furthermore, NLRP3 deficiency impaired the migration of neutrophils and chemokine expression by macrophages. These findings demonstrate that NLRP3 regulates Stat3 signaling in alveolar epithelial cells by affecting macrophage and neutrophil function independent of IL-1ß production and contributes to the pathophysiology of HALI.


Subject(s)
Acute Lung Injury/metabolism , Carrier Proteins/genetics , Hyperoxia/metabolism , Interleukin-1beta/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Carrier Proteins/metabolism , Hyperoxia/genetics , Hyperoxia/pathology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Proto-Oncogene Proteins c-bcl-2/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
17.
Heart Vessels ; 29(6): 867-72, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24281399

ABSTRACT

We describe three cases of J-wave syndrome in which ventricular fibrillation (VF) was probably induced by corticosteroid therapy. The patients involved were being treated with prednisolone for concomitant bronchial asthma. One of the three patients had only one episode of VF during her long follow-up period (14 years). Two patients had hypokalemia during their VF episodes. Corticosteroids have been shown to induce various types of arrhythmia and to modify cardiac potassium channels. We discuss the possible association between corticosteroid therapy and VF in J-wave syndrome based on the cases we have encountered.


Subject(s)
Anti-Arrhythmia Agents/administration & dosage , Cardiopulmonary Resuscitation/methods , Defibrillators, Implantable , Glucocorticoids/adverse effects , Hypokalemia , Ventricular Fibrillation , Adult , Asthma/drug therapy , Electrocardiography/methods , Female , Glucocorticoids/administration & dosage , Humans , Hypokalemia/chemically induced , Hypokalemia/complications , Hypokalemia/diagnosis , Male , Syndrome , Treatment Outcome , Ventricular Fibrillation/diagnosis , Ventricular Fibrillation/etiology , Ventricular Fibrillation/therapy
18.
Hypertens Res ; 35(4): 426-33, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22129515

ABSTRACT

Fetuin-A and osteoprotegerin (OPG) are arterial calcification regulators, which are related to cardiovascular survival in hemodialysis patients. We hypothesized that a balance of these calcification regulators might mediate the progression of left ventricular (LV) diastolic dysfunction in hemodialysis patients. We recruited 63 hemodialysis patients and measured their serum fetuin-A, OPG, arterial stiffness, aortic calcification and echocardiographic parameters, including the transmitral early diastolic velocity/tissue Doppler mitral annular early diastolic velocity ratio (E/E'), and analyzed the relationships between these variables. Fetuin-A levels were significantly and negatively correlated with the ankle-brachial pulse wave velocity (baPWV), aortic calcification score (AOCS), left atrial volume index (LAVI), LV mass index (LVMI) and E/E'. OPG levels and the ratio of OPG to fetuin-A levels were significantly and positively correlated with the baPWV, AOCS, LAVI and E/E'. A stepwise multiple regression analysis revealed that E/E' was independently correlated with fetuin-A levels (ß=-0.334, P=0.02), OPG levels (ß=0.367, P=0.01) and the ratio of OPG to fetuin-A (ß=0.295, P=0.04). Categorizing the patients according to their serum fetuin-A and OPG levels revealed that patients with low fetuin-A and high OPG levels had the highest LAVI, LVMI and E/E' values after adjusting for potential confounders. Serum fetuin-A levels negatively reflected, whereas OPG levels and the ratio of OPG to fetuin-A positively reflected an increase in vascular and ventricular stiffness, leading to the aggravation of diastolic dysfunction. Therefore, based on our results, the balance of the tissue calcification regulators fetuin-A and OPG could mediate the progression of LV diastolic dysfunction in hemodialysis patients.


Subject(s)
Osteoprotegerin/blood , Renal Dialysis , Renal Insufficiency/blood , Ventricular Dysfunction, Left/blood , alpha-2-HS-Glycoprotein/metabolism , Aged , Ankle Brachial Index , Blood Flow Velocity/physiology , Diastole/physiology , Female , Humans , Male , Middle Aged , Renal Insufficiency/complications , Renal Insufficiency/physiopathology , Renal Insufficiency/therapy , Ventricular Dysfunction, Left/complications , Ventricular Dysfunction, Left/physiopathology
19.
Int Heart J ; 52(5): 308-11, 2011.
Article in English | MEDLINE | ID: mdl-22008442

ABSTRACT

Fabry disease is an X-linked lysosomal storage disorder caused by mutations of the α-galactosidase A gene (GLA), and the disease is a relatively prevalent cause of left ventricular hypertrophy mimicking idiopathic hypertrophic cardiomyopathy. We assessed clinically 5 patients of a three-generation family and also searched for GLA mutations in 10 family members. The proband had left ventricular hypertrophy with localized thinning in the basal posterior wall and late gadolinium enhancement (LGE) in the near-circumferential wall in cardiovascular magnetic resonance images and her sister had vasospastic angina pectoris without organic stenosis of the coronary arteries. LGE notably appeared in parallel with decreased α-galactosidase A activity and increased NT-pro BNP in our patients. We detected a new GLA missense mutation (G195V) in exon 4, resulting in a glycine-to-valine substitution. Of the 10 family members, 5 family members each were positive and negative for this mutation. These new data extend our clinical and molecular knowledge of GLA gene mutations and confirm that a novel missense mutation in the GLA gene is important not only for a precise diagnosis of heterozygous status, but also for confirming relatives who are negative for this mutation.


Subject(s)
Fabry Disease/diagnosis , Fabry Disease/genetics , Hypertrophy, Left Ventricular/diagnosis , Hypertrophy, Left Ventricular/genetics , Mutation, Missense/genetics , alpha-Galactosidase/genetics , Adult , Amino Acid Substitution/genetics , Bundle-Branch Block/diagnosis , Bundle-Branch Block/genetics , Bundle-Branch Block/pathology , Coronary Angiography , Coronary Vasospasm/diagnosis , Coronary Vasospasm/genetics , Coronary Vasospasm/pathology , DNA Mutational Analysis , Echocardiography , Electrocardiography , Exons/genetics , Fabry Disease/pathology , Female , Genotype , Glycine/genetics , Humans , Hypertrophy, Left Ventricular/pathology , Japan , Magnetic Resonance Imaging , Male , Microscopy, Electron , Middle Aged , Myocardium/pathology , Natriuretic Peptide, Brain/blood , Pedigree , Peptide Fragments/blood , Signal Processing, Computer-Assisted , Valine/genetics , Young Adult
20.
Hypertens Res ; 31(2): 193-201, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18360037

ABSTRACT

Chronic kidney disease (CKD) is associated with an increased risk of cardiovascular disease, and thus is a major worldwide public health problem. Recently, an estimated glomerular filtration rate (eGFR) using the Modification of Diet in Renal Disease equation for Japanese patients was proposed by the Japanese Society of Nephrology. However, the role of eGFR in the assessment of atherosclerosis is not well understood in Japanese patients. We analyzed the relationship between eGFR and severity of arterial stiffness using brachial-ankle pulse wave velocity (baPWV) in 647 adult Japanese patients. baPWV correlated significantly and positively with age, hypertension, diabetes, prior cardiovascular disease, blood pressure, pulse pressure and heart rate, and negatively with eGFR (r=-0.405, p<0.0001). A multiple regression analysis revealed that baPWV correlated independently with eGFR. Furthermore, there was a stepwise increase in baPWV, corresponding to advances in CKD through stages 1 to 5. When CKD stage 3 was divided at eGFR 45 mL/min/1.73 m2, the baPWV of stage 3b (eGFR 30 to 44) was significantly higher than that of stage 3a (eGFR 45 to 59) independent of traditional risk factors, suggesting that an eGFR of 45 mL/min/1.73 m2 may be a critical cut off value to predict arterial stiffness in CKD. In conclusion, the newly proposed eGFR is significantly associated with arterial stiffness, independent of traditional risk factors for cardiovascular disease.


Subject(s)
Arteries/physiopathology , Glomerular Filtration Rate , Kidney Diseases/physiopathology , Adolescent , Adult , Aged , Aged, 80 and over , Atherosclerosis/etiology , Blood Flow Velocity , Body Mass Index , Brachial Artery , Cardiovascular Diseases/etiology , Chronic Disease , Female , Humans , Male , Middle Aged , Pulsatile Flow , Regression Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...