Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Kurume Med J ; 63(1.2): 33-37, 2017 Apr 13.
Article in English | MEDLINE | ID: mdl-28090005

ABSTRACT

Because of the protective effect of estrogen for atherosclerosis, the prevalence of acute coronary syndrome in women before menopause is low. We report a rare case of unstable angina in a young Japanese female who had a history of cigarette smoking and contraceptive use. Her coronary stenosis was successfully treated by percutaneous coronary intervention.


Subject(s)
Angina, Unstable/diagnosis , Percutaneous Coronary Intervention/methods , Angina, Unstable/surgery , Contraceptives, Oral/adverse effects , Coronary Angiography , Coronary Artery Disease/diagnosis , Coronary Artery Disease/surgery , Female , Humans , Japan , Smoking , Young Adult
3.
PLoS One ; 10(5): e0127942, 2015.
Article in English | MEDLINE | ID: mdl-26010537

ABSTRACT

Myocardial ischemia reperfusion injury (IRI) adversely affects cardiac performance and the prognosis of patients with acute myocardial infarction. Although myocardial signal transducer and activator of transcription (STAT) 3 is potently cardioprotective during IRI, the inhibitory mechanism responsible for its activation is largely unknown. The present study aimed to investigate the role of the myocardial suppressor of cytokine signaling (SOCS)-3, an intrinsic negative feedback regulator of the Janus kinase (JAK)-STAT signaling pathway, in the development of myocardial IRI. Myocardial IRI was induced in mice by ligating the left anterior descending coronary artery for 1 h, followed by different reperfusion times. One hour after reperfusion, the rapid expression of JAK-STAT-activating cytokines was observed. We precisely evaluated the phosphorylation of cardioprotective signaling molecules and the expression of SOCS3 during IRI and then induced myocardial IRI in wild-type and cardiac-specific SOCS3 knockout mice (SOCS3-CKO). The activation of STAT3, AKT, and ERK1/2 rapidly peaked and promptly decreased during IRI. This decrease correlated with the induction of SOCS3 expression up to 24 h after IRI in wild-type mice. The infarct size 24 h after reperfusion was significantly reduced in SOCS3-CKO compared with wild-type mice. In SOCS3-CKO mice, STAT3, AKT, and ERK1/2 phosphorylation was sustained, myocardial apoptosis was prevented, and the expression of anti-apoptotic Bcl-2 family member myeloid cell leukemia-1 (Mcl-1) was augmented. Cardiac-specific SOCS3 deletion led to the sustained activation of cardioprotective signaling molecules including and prevented myocardial apoptosis and injury during IRI. Our findings suggest that SOCS3 may represent a key factor that exacerbates the development of myocardial IRI.


Subject(s)
MAP Kinase Signaling System , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Gene Deletion , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardium/pathology , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics
4.
Circ J ; 79(7): 1557-67, 2015.
Article in English | MEDLINE | ID: mdl-25833080

ABSTRACT

BACKGROUND: Remote ischemic preconditioning (RIPC) induced by transient limb ischemia is a powerful innate mechanism of cardioprotection against ischemia. Several described mechanisms explain how RIPC may act through neural pathways or humoral factors; however, the mechanistic pathway linking the remote organ to the heart has not yet been fully elucidated. This study aimed to investigate the mechanisms underlying the RIPC-induced production of Janus kinase (JAK)-signal transducer and activator of the transcription (STAT)-activating cytokines and cardioprotection by using mouse and human models of RIPC. METHODS AND RESULTS: Screened circulating cardioprotective JAK-STAT-activating cytokines in mice unexpectedly revealed increased serum erythropoietin (EPO) levels after RIP induced by transient ischemia. In mice, RIPC rapidly upregulated EPO mRNA and its main transcriptional factor, hypoxia-inducible factor-1α (HIF1α), in the kidney. Laser Doppler blood flowmetry revealed a prompt reduction of renal blood flow (RBF) after RIPC. RIPC activated cardioprotective signaling pathways and the anti-apoptotic Bcl-xL pathway in the heart, and reduced infarct size. In mice, these effects were abolished by administration of an EPO-neutralizing antibody. Renal nerve denervation also abolished RIPC-induced RBF reduction, EPO production, and cardioprotection. In humans, transient limb ischemia of the upper arm reduced RBF and increased serum EPO levels. CONCLUSIONS: Based on the present data, we propose a novel RIPC mechanism in which inhibition of infarct size by RIPC is produced through the renal nerve-mediated reduction of RBF associated with activation of the HIF1α-EPO pathway.


Subject(s)
Erythropoietin/metabolism , Ischemic Preconditioning, Myocardial , Kidney/innervation , Myocardial Infarction/metabolism , Peripheral Nerves/metabolism , Animals , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Janus Kinases/metabolism , Kidney/blood supply , Kidney/metabolism , Kidney/physiopathology , Male , Mice , Mice, Inbred BALB C , Myocardial Infarction/physiopathology , Myocardial Infarction/prevention & control , Peripheral Nerves/physiopathology , bcl-X Protein/metabolism
5.
J Am Coll Cardiol ; 63(24): 2722-33, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24768874

ABSTRACT

OBJECTIVES: This study examined the impact of omentin on myocardial injury in a mouse model of ischemia/reperfusion (I/R) and explored its underlying mechanisms. BACKGROUND: Obesity is a major risk factor for ischemic heart disease. Omentin is a circulating adipokine that is down-regulated by obesity. METHODS: In patients who underwent successful reperfusion treatment after acute myocardial infarction, cardiac function and perfusion defect were assessed by using scintigraphic images. Mice were subjected to myocardial ischemia followed by reperfusion. RESULTS: This study found that high levels of plasma omentin were associated with improvement of heart damage and function after reperfusion therapy in patients with acute myocardial infarction. Systemic administration of human omentin to mice led to a reduction in myocardial infarct size and apoptosis after I/R, which was accompanied by enhanced phosphorylation of AMP-activated protein kinase (AMPK) and Akt in the ischemic heart. Fat-specific overexpression of human omentin also resulted in reduction of infarct size after I/R. Blockade of AMPK or Akt activity reversed omentin-induced inhibition of myocardial ischemic damage and apoptosis in mice. In cultured cardiomyocytes, omentin suppressed hypoxia/reoxygenation-induced apoptosis, which was blocked by inactivation of AMPK or Akt. CONCLUSIONS: Our data indicate that omentin functions as an adipokine that ameliorates acute ischemic injury in the heart by suppressing myocyte apoptosis through both AMPK- and Akt-dependent mechanisms.


Subject(s)
AMP-Activated Protein Kinases/physiology , Cytokines/therapeutic use , Lectins/therapeutic use , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/prevention & control , Oncogene Protein v-akt/physiology , Animals , Apoptosis/genetics , Biomarkers/blood , Cytokines/blood , Cytokines/genetics , Disease Models, Animal , Female , GPI-Linked Proteins/blood , GPI-Linked Proteins/genetics , GPI-Linked Proteins/therapeutic use , Humans , Lectins/blood , Lectins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Myocardial Reperfusion/methods , Myocardial Reperfusion Injury/pathology , Percutaneous Coronary Intervention/methods , Phosphorylation/genetics
9.
Int J Cardiol ; 160(3): 216-9, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-22841473

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are highly potent professional antigen-presenting cells that play a central role in initiating the primary immune response. Accumulating evidence suggests that immune-mediated inflammation plays an important role in the pathophysiology of AMI, but the mechanism that triggers such immune responses is unknown. METHODS: Using multi-color flow-cytometry, we determined the numbers of circulating myeloid DCs (mDCs) and plasmacytoid DCs (pDCs) in patients with AMI (n = 26) or stable angina pectoris (SAP) (n = 19), and in age-matched control subjects (n = 19). The DC activation markers CD40 and CD83 were also measured. RESULTS: On admission, circulating mDC and pDC counts were significantly lower in AMI patients compared to control subjects and SAP patients (mDC, P < 0.01; pDC, P < 0.05). The activation markers of mDCs in AMI patients were significantly higher and returned to the levels of control subjects or SAP patients 3 days after AMI (mDC, P < 0.05; pDC, P < 0.05). Reductions of circulating mDC and pDC numbers were restored 7 days after the onset of AMI. Furthermore, we found that the recovery of the circulating DC numbers 14 days after AMI was correlated with the alterations of creatine kinase-MB (CK-MB) (mDC, r = 0.48, P < 0.05; pDC, r=0.52, P < 0.01) and brain natriuretic peptide (BNP) (mDC, r = 0.53, P < 0.01; pDC, r = 0.51, P < 0.01). CONCLUSION: Our findings suggest that the transient reduction and activation of circulating DCs may play important roles in the pathophysiology of myocardial injury after AMI.


Subject(s)
Dendritic Cells/metabolism , Flow Cytometry/methods , Myocardial Infarction/blood , Aged , Aged, 80 and over , Cell Count/methods , Female , Humans , Male , Middle Aged , Myocardial Infarction/diagnosis , Time Factors
10.
Hypertens Res ; 35(11): 1063-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22763477

ABSTRACT

Lysyl oxidase (LOX) and LOX-like protein-1 (LOXL-1) are extracellular matrix-embedded amine oxidases that have critical roles in the cross-linking of collagen and elastin. LOX family proteins are abundantly expressed in the remodeled heart of animals and humans and are implicated in cardiac fibrosis; however, their role in cardiac hypertrophy is unknown. In this study, in vitro stimulation with hypertrophic agonists significantly increased LOXL-1 expression, LOX enzyme activity and [(3)H] leucine incorporation in neonatal rat cardiomyocytes. A LOX inhibitor, beta-aminopropionitrile (BAPN), inhibited agonist-induced leucine incorporation in cardiomyocytes in vitro, suggesting the involvement of LOXL-1 in cardiomyocyte hypertrophy. Abdominal aortic constriction in rats produced left ventricular hypertrophy in parallel with LOXL-1 mRNA upregulation. And BAPN administration significantly inhibited angiotensin II-induced cardiac hypertrophy in vivo. These results suggest a role of LOXL-1 in cardiac hypertrophy in vivo. We generated transgenic mice with cardiomyocyte-specific expression of LOXL-1. LOXL-1 transgenic mice pups were born normally and grew to adulthood without increased mortality; these mice exhibited a greater left ventricle to body weight ratio, larger myocyte diameter, and more brain natriuretic peptide expression than their wild-type littermates. Echocardiography revealed that the LOXL-1 transgenic mice also had greater wall thickness with preserved cardiac contraction. Our results indicate a possible fundamental role of LOXL-1 in cardiac hypertrophy.


Subject(s)
Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Myocytes, Cardiac/metabolism , Amino Acid Oxidoreductases/antagonists & inhibitors , Aminopropionitrile/pharmacology , Angiotensin II/adverse effects , Angiotensin II/drug effects , Angiotensin II/pharmacology , Animals , Animals, Newborn , Cardiomegaly/chemically induced , Cells, Cultured , Disease Models, Animal , Echocardiography , Endothelin-1/pharmacology , In Vitro Techniques , Leucine/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Rats , Rats, Wistar
11.
J Am Coll Cardiol ; 59(9): 838-52, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22361405

ABSTRACT

OBJECTIVES: The study investigated the role of myocardial suppressor of cytokine signaling-3 (SOCS3), an intrinsic negative feedback regulator of the janus kinase and signal transducer and activator of transcription (JAK-STAT) signaling pathway, in the development of left ventricular (LV) remodeling after acute myocardial infarction (AMI). BACKGROUND: LV remodeling after AMI results in poor cardiac performance leading to heart failure. Although it has been shown that JAK-STAT-activating cytokines prevent LV remodeling after AMI in animals, little is known about the role of SOCS3 in this process. METHODS: Cardiac-specific SOCS3 knockout mice (SOCS3-CKO) were generated and subjected to AMI induced by permanent ligation of the left anterior descending coronary artery. RESULTS: Although the initial infarct size after coronary occlusion measured by triphenyltetrazolium chloride staining was comparable between SOCS3-CKO and control mice, the infarct size 14 days after AMI was remarkably inhibited in SOCS3-CKO, indicating that progression of LV remodeling after AMI was prevented in SOCS3-CKO hearts. Prompt and marked up-regulations of multiple JAK-STAT-activating cytokines including leukemia inhibitory factor and granulocyte colony-stimulating factor (G-CSF) were observed within the heart following AMI. Cardiac-specific SOCS3 deletion enhanced multiple cardioprotective signaling pathways including STAT3, AKT, and extracellular signal-regulated kinase (ERK)-1/2, while inhibiting myocardial apoptosis and fibrosis as well as augmenting antioxidant expression. CONCLUSIONS: Enhanced activation of cardioprotective signaling pathways by inhibiting myocardial SOCS3 expression prevented LV remodeling after AMI. Our data suggest that myocardial SOCS3 may be a key molecule in the development of LV remodeling after AMI.


Subject(s)
Gene Deletion , Myocardial Infarction/genetics , Myocardium/metabolism , RNA/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Ventricular Dysfunction, Left/etiology , Ventricular Remodeling/genetics , Animals , Blotting, Western , Disease Models, Animal , Disease Progression , Echocardiography , Heart Ventricles/diagnostic imaging , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred BALB C , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , Up-Regulation , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology
12.
JAKSTAT ; 1(4): 234-40, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-24058778

ABSTRACT

The suppressors of cytokine signaling (SOCS) family of proteins are cytokine-inducible inhibitors of Janus kinase (JAK)-signal transducer and activator of the transcription (STAT) signaling pathways. Among the family, SOCS1 and SOCS3 potently suppress cytokine actions by inhibiting JAK kinase activities. The generation of mice lacking individual SOCS genes has been instrumental in defining the role of individual SOCS proteins in specific cytokine pathways in vivo; SOCS1 is an essential negative regulator of interferon-γ (IFNγ) and SOCS3 is an essential negative regulator of leukemia inhibitory factor (LIF). JAK-STAT3 activating cytokines have exhibited cardioprotective roles in the heart. The cardiac-specific deletion of SOCS3 enhances the activation of cardioprotective signaling pathways, inhibits myocardial apoptosis and fibrosis and results in the inhibition of left ventricular remodeling after myocardial infarction (MI). We propose that myocardial SOCS3 is a key determinant of left ventricular remodeling after MI, and SOCS3 may serve as a novel therapeutic target to prevent left ventricular remodeling after MI. In this review, we discuss the signaling pathways mediated by JAK-STAT and SOCS proteins and their roles in the development of myocardial injury under stress (e.g., pressure overload, viral infection and ischemia).

14.
JACC Cardiovasc Imaging ; 4(10): 1110-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21999871

ABSTRACT

OBJECTIVES: The aim of this study was to compare the effect of pioglitazone, an insulin sensitizer, with glimepiride, an insulin secretagogue, on atherosclerotic plaque inflammation by using serial (18)F-fluorodeoxyglucose positron emission tomography (FDG-PET) imaging. BACKGROUND: Atherosclerosis is intrinsically an inflammatory disease. Although hyperglycemia is associated with an increased risk of atherosclerotic cardiovascular disease, there are no clinical data to show the preference of any specific oral hypoglycemic agents to prevent atherosclerotic plaque inflammation. METHODS: A total of 56 impaired glucose tolerant or diabetic patients with carotid atherosclerosis underwent a complete history, determinations of blood chemistries, anthropometric variables, and FDG-PET. They were randomly assigned to receive either pioglitazone (15 to 30 mg) or glimepiride (0.5 to 4.0 mg) for 4 months with titration to optimal dosage. Effects of the drugs on atherosclerotic plaque inflammation were evaluated by FDG-PET at study completion. Plaque inflammation was measured by blood-normalized standardized uptake value, known as a target-to-background ratio. RESULTS: The study was completed in 31 pioglitazone-treated patients and 21 glimepiride-treated patients. Although both treatments reduced fasting plasma glucose and hemoglobin A1c values comparably, pioglitazone, but not glimepiride, decreased atherosclerotic plaque inflammation. Compared with glimepiride, pioglitazone significantly increased high-density lipoprotein cholesterol level. High-sensitivity C-reactive protein was decreased by pioglitazone, whereas it was increased by glimepiride. Multiple stepwise regression analysis revealed that the increase in high-density lipoprotein cholesterol level was independently associated with the attenuation of plaque inflammation. CONCLUSIONS: Our present study suggests that pioglitazone could attenuate atherosclerotic plaque inflammation in patients with impaired glucose tolerance or in diabetic patients independent of glucose lowering effect. Pioglitazone may be a promising strategy for the treatment of atherosclerotic plaque inflammation in impaired glucose tolerance or diabetic patients. (Detection of Plaque Inflammation and Visualization of Anti-Inflammatory Effects of Pioglitazone on Plaque Inflammation in Subjects With Impaired Glucose Tolerance and Type 2 Diabetes Mellitus by FDG-PET/CT; NCT00722631).


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Aortic Diseases/drug therapy , Aortography , Carotid Artery Diseases/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/drug therapy , Fluorodeoxyglucose F18 , Glucose Intolerance/drug therapy , Hypoglycemic Agents/therapeutic use , Inflammation/drug therapy , Multimodal Imaging , Positron-Emission Tomography , Radiopharmaceuticals , Sulfonylurea Compounds/therapeutic use , Thiazolidinediones/therapeutic use , Tomography, X-Ray Computed , Aged , Aortic Diseases/diagnostic imaging , Aortic Diseases/etiology , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , C-Reactive Protein/metabolism , Carotid Artery Diseases/diagnostic imaging , Carotid Artery Diseases/etiology , Cholesterol, HDL/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetic Angiopathies/diagnostic imaging , Diabetic Angiopathies/etiology , Female , Glucose Intolerance/blood , Glucose Intolerance/complications , Glycated Hemoglobin/metabolism , Humans , Inflammation/diagnostic imaging , Inflammation/etiology , Japan , Male , Middle Aged , Pioglitazone , Predictive Value of Tests , Prospective Studies , Regression Analysis , Time Factors , Treatment Outcome
15.
Int J Cardiol ; 147(2): 258-64, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-19923020

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are the most potent antigen-presenting cells and play a central role in initiating the primary immune response. Although increasing evidence supports immune-mediated inflammation plays an important role in the pathophysiology of heart failure, little is known regarding the source and mechanism that trigger immune responses. The present study examined whether circulating DCs have any role in the pathophysiology in heart failure in humans. METHODS AND RESULTS: With multi-color flow cytometry we determined the numbers of circulating myeloid DCs (mDCs) and plasmacytoid DCs (pDCs) in decompensated heart failure patients with NYHA class III or IV on admission (n = 27) and the age-similar control subjects (n = 21). DC activation markers such as CD40, and CCR7 were also measured. On admission, circulating mDC and pDC counts were significantly lower in decompensated heart failure patients compared to control subjects (p < 0.01). Circulating mDCs and pDCs were activated in the decompensated heart failure patients. Heart failure treatment restored the reduction and the activation of circulating mDCs and pDCs (p < 0.05). The increases of circulating DCs numbers after treatment were correlated with the decreases in B-type natriuretic peptide (BNP) and troponin-T (p < 0.05) and with the increase in left ventricular ejection fraction (LVEF) (p < 0.01). Furthermore, we found that poor recovery of the circulating DCs number after treatment predicted recurrence of decompensated heart failure. CONCLUSION: These findings suggest that the reduction and activation of circulating DCs may be involved in the pathophysiology of heart failure.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/immunology , Heart Failure/immunology , Heart Failure/pathology , Aged , Cell Count , Female , Flow Cytometry/methods , Fluorescent Antibody Technique , Heart Failure/physiopathology , Humans , Male , Middle Aged , Predictive Value of Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...