Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Intern Med ; 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38008457

ABSTRACT

Gastroduodenal stenting (GDS) is a less invasive alternative to gastrojejunostomy for the management of malignant gastric outlet obstruction (mGOO). GDS is a minimally invasive treatment with good technical and clinical success, and severe complications that require surgical intervention are rare. Stent fracture is an uncommon complication associated with GDS; however, migration of the fractured distal segment can result in small bowel obstruction. Adverse effects of stent fractures in patients with mGOO have rarely been reported. We herein report two surgical cases of small bowel obstruction caused by the migration of fractured metal stent in patients with mGOO.

2.
Ann Cardiothorac Surg ; 7(1): 99-105, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29492387

ABSTRACT

BACKGROUND: Previously, we have demonstrated the cardioprotective effect of interleukin (IL)-11 in animal models of acute coronary syndrome. In this study, we sought to evaluate its cardioprotective potential during prolonged hypothermic global ischemia and subsequent reperfusion using a rat heart donor model. METHODS: IL-11 was administered intravenously 10 minutes before harvesting the rat heart. The hearts were preserved in cold (4 °C) Krebs-Henseleit buffer for 6 hours, and then attached to a Langendorff perfusion apparatus and reperfused with an oxygenated Krebs-Henseleit solution containing IL-11. Normal saline was used instead of IL-11 in the control group. Functional recovery of the reperfused heart was observed by using a left ventricular balloon. Myocardial cell injury was quantified by measuring the biomarkers collected from the coronary effluent. Apoptotic cells were identified and counted using the terminal deoxynucleotide transferase-mediated dUTP nick-end labeling (TUNEL) staining technique. RESULTS: IL-11 administration improved myocardial function after 6 hours of cold ischemia. Although there were no significant differences in any of the baseline-measured values between the two groups, left ventricular developed pressure (LVDP) and changes in left ventricular pressures (dP/dt) were significantly higher in the IL-11 group at 120-minute reperfusion. The number of TUNEL-labeled cardiomyocytes was also significantly smaller in the IL-11 group. CONCLUSIONS: The administration of IL-11 showed a significant recovery of cardiac contractile function after 6 hours of cold ischemia. Our data suggest that it may have significant therapeutic potential for maintaining the functional viability of the heart exposed to prolonged hypothermic global ischemia.

3.
J Cardiovasc Pharmacol Ther ; 22(3): 273-282, 2017 05.
Article in English | MEDLINE | ID: mdl-27811197

ABSTRACT

BACKGROUND: Phospholamban (PLN) inhibition enhances calcium cycling and is a potential novel therapy for heart failure (HF). Antisense oligonucleotides (ASOs) are a promising tool for unmet medical needs. Nonviral vector use of locked nucleic acid (LNA)-modified ASOs (LNA-ASOs), which shows strong binding to target RNAs and is resistant to nuclease, is considered to have a potential for use in novel therapeutics in the next decades. Thus, the efficacy of a single-dose injection of LNA-ASO for cardiac disease needs to be elucidated. We assessed the therapeutic efficacy of a single-dose LNA-ASO injection targeting PLN in pressure overload-induced cardiac dysfunction. METHODS AND RESULTS: Mice intravenously injected with Cy3-labeled LNA-ASO displayed Cy3 fluorescence in the liver and heart 24 hours after injection. Subsequently, male C57BL/6 mice were subjected to sham or transverse aortic constriction surgery; after 3 weeks, these were treated with PLN-targeting LNA-ASO (0.3 mg/kg) or scrambled LNA-ASO. Cardiac function was measured by echocardiography before and 1 week after injection. Phospholamban-targeting LNA-ASO treatment significantly improved fractional shortening (FS) by 6.5%, whereas administration of the scrambled LNA-ASO decreased FS by 4.0%. CONCLUSION: Our study revealed that a single-dose injection of PLN-targeting LNA-ASO improved contractility in pressure overload-induced cardiac dysfunction, suggesting that LNA-ASO is a promising tool for hypertensive HF treatment.


Subject(s)
Calcium-Binding Proteins/metabolism , Heart Failure/drug therapy , Myocardium/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides/administration & dosage , Systole/drug effects , Ventricular Dysfunction, Left/drug therapy , Ventricular Function, Left/drug effects , Animals , Blood Pressure , Calcium-Binding Proteins/genetics , Disease Models, Animal , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Hepatocytes/metabolism , Hypertension/physiopathology , Hypertrophy, Left Ventricular/drug therapy , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Injections, Intravenous , Male , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oligonucleotides/genetics , Oligonucleotides, Antisense/genetics , Recovery of Function , Time Factors , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
4.
World J Surg Oncol ; 14(1): 170, 2016 Jun 27.
Article in English | MEDLINE | ID: mdl-27349744

ABSTRACT

BACKGROUND: Prognostic nutritional index has been shown to be a prognostic marker for various solid tumors. However, few studies have investigated the impact of the prognostic nutritional index on survival of patients with breast cancer. The aim of this study was to investigate the impact of the prognostic nutritional index on the long-term outcomes in patients with breast cancer. METHODS: This study reviewed the medical records of 212 patients with breast cancer who underwent mastectomy. The prognostic nutritional index was calculated as 10 × serum albumin (g/dl) + 0.005 × total lymphocyte count (per mm(3)). Receiver operating characteristic curve analysis was performed to determine the cutoff value of the prognostic nutritional index. The survival curves were calculated by the Kaplan-Meier method. Differences between the curves were analyzed by the log-rank test. Multivariate Cox proportional hazard model was used to evaluate the prognostic significance of prognostic nutritional index in patients with breast cancer. RESULTS: The mean prognostic nutritional index just before the operation was 51.9, and the median follow-up after surgery was 47.7 months. The optimal cutoff value of the prognostic nutritional index for predicting the overall survival was 52.8 from the receiver operating characteristic curve analysis. The 5-year overall survival rate was 98.3 % in the prognostic nutritional index >52.8 and 92.0 % in the prognostic nutritional index <52.8 (P = 0.013). In the multivariate analysis, a low prognostic nutritional index was an independent predictor for poor overall survival (HR, 5.88; 95 % CI, 1.13-108.01; P = 0.033). CONCLUSIONS: The prognostic nutritional index is a simple and useful marker for predicting the long-term outcomes of breast cancer patients, independent of the tumor stage.


Subject(s)
Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/therapy , Carcinoma, Lobular/therapy , Nutritional Status , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/pathology , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Middle Aged , Multivariate Analysis , Neoplasm Staging , Nutrition Assessment , Prognosis , ROC Curve , Retrospective Studies , Survival Rate
5.
Cytokine ; 75(2): 365-72, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26142823

ABSTRACT

Cytokines play important roles in cardiac repair and regeneration. Recently, we demonstrated that interleukin (IL)-6 family cytokines induce the endothelial differentiation of Sca-1+ cardiac resident stem cells through STAT3/Pim-1 signaling pathway. In contrast, the biological functions of IL-12 family cytokines in heart remain to be elucidated, though they show structural homology with IL-6. In the present study, we examined the effects of IL-12 family cytokines on the transdifferentiation of cardiac Sca-1+ cells into cardiac cells. RT-PCR analyses revealed that IL-27 receptor α (IL-27Rα), but not IL-12R or IL-23R, was expressed in cardiac Sca-1+ cells. The transcript expression of IL-27 was elevated in murine hearts in cardiac injury models. Intriguingly, IL-27 stimulation for 14 days induced the endothelial cell (EC) marker genes, such as CD-31 and VE-cadherin. Immunoblot analyses clarified that IL-27 treatment rapidly phosphorylated STAT3. IL-27 upregulated the expression of Pim-1, but the overexpression of dominant negative STAT3 abrogated the induction of Pim-1 by IL-27. Finally, adenoviral transfection of dominant negative Pim-1 inhibited IL-27-induced EC differentiation of cardiac Sca-1+ cells. These findings demonstrated that IL-27 promoted the commitment of cardiac stem cells into the EC lineage, possibly leading to neovascularization as a novel biological function. IL-27 could not only regulate the inflammation but also contribute to the maintenance of the tissue homeostasis through stem cell differentiation at inflammatory sites.


Subject(s)
Interleukins/pharmacology , Myocardium/cytology , Proto-Oncogene Proteins c-pim-1/metabolism , STAT3 Transcription Factor/metabolism , Stem Cells/cytology , Animals , Cadherins/biosynthesis , Cell Differentiation/physiology , Cell Transdifferentiation/physiology , Cells, Cultured , Endothelial Cells/cytology , Heart Injuries/pathology , Interleukin-12/immunology , Interleukins/immunology , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Proto-Oncogene Proteins c-pim-1/biosynthesis , Receptors, Cytokine/biosynthesis , Receptors, Interleukin/biosynthesis , Receptors, Interleukin-12/biosynthesis
6.
PLoS One ; 9(10): e111097, 2014.
Article in English | MEDLINE | ID: mdl-25340554

ABSTRACT

Interleukin-6 (IL-6) family cytokines play important roles in cardioprotection against pathological stresses. IL-6 cytokines bind to their specific receptors and activate glycoprotein 130 (gp130), a common receptor, followed by further activation of STAT3 and extracellular signal-regulated kinase (ERK)1/2 through janus kinases (JAKs); however the importance of glycosylation of gp130 remains to be elucidated in cardiac myocytes. In this study, we examined the biological significance of gp130 glycosylation using tunicamycin (Tm), an inhibitor of enzyme involved in N-linked glycosylation. In cardiomyocytes, the treatment with Tm completely replaced the glycosylated form of gp130 with its unglycosylated one. Tm treatment inhibited leukemia inhibitory factor (LIF)-mediated activation of STAT3 and ERK1/2. Similarly, IL-11 failed to activate STAT3 and ERK1/2 in the presence of Tm. Interestingly, Tm inhibited the activation of JAKs 1 and 2, without influencing the expression of suppressor of cytokine signalings (SOCSs) and protein-tyrosine phosphatase 1B (PTP1B), which are endogenous inhibitors of JAKs. To exclude the possibility that Tm blocks LIF and IL-11 signals by inhibiting the glycosylation of their specific receptors, we investigated whether the stimulation with IL-6 plus soluble IL-6 receptor (sIL-6R) could transduce their signals in Tm-treated cardiomyocytes and found that this stimulation was unable to activate the downstream signals. Collectively, these findings indicate that glycosylation of gp130 is essential for signal transduction of IL-6 family cytokines in cardiomyocytes.


Subject(s)
Cytokine Receptor gp130/metabolism , Interleukin-6/chemistry , Myocytes, Cardiac/metabolism , STAT3 Transcription Factor/metabolism , Tunicamycin/chemistry , Animals , Animals, Newborn , Cells, Cultured , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycosylation , Microscopy, Fluorescence , Rats , Receptors, Interleukin-6/chemistry , Signal Transduction
7.
PLoS One ; 9(10): e110483, 2014.
Article in English | MEDLINE | ID: mdl-25334017

ABSTRACT

SLD5 is a member of the GINS complex composed of PSF1, PSF2, PSF3 and SLD5, playing a critical role in the formation of the DNA replication fork with CDC45 in yeast. Previously, we had isolated a PSF1 orthologue from a murine hematopoietic stem cell DNA library and were then able to identify orthologues of all the other GINS members by the yeast two hybrid approach using PSF1 as the bait. These GINS orthologues may also function in DNA replication in mammalian cells because they form tetrameric complexes as observed in yeast, and gene deletion mutants of both PSF1 and SLD5 result in a lack of epiblast proliferation and early embryonic lethality. However, we found that PSF1 is also involved in chromosomal segregation in M phase, consistent with recent suggestions that homologues of genes associated with DNA replication in lower organisms also regulate cellular events other than DNA replication in mammalian cells. Here we analyzed the function of SLD5 other than DNA replication and found that it is active in DNA damage and repair. Attenuation of SLD5 expression results in marked DNA damage in both normal cells and cancer cells, suggesting that it protects against DNA damage. Attenuation of SLD5 delays the DNA repair response and cell cycle restoration in normal cells but not in cancer cells. These findings suggest that SLD5 might represent a therapeutic target molecule acting at the level of tumor stromal cells rather than the cancerous cells themselves, because development of the tumor microenvironment could be delayed or disrupted by the suppression of its expression in the normal cell types within the tumor.


Subject(s)
Carrier Proteins/metabolism , DNA Damage , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Cycle Checkpoints , Cell Line , DNA Repair , Mice , RNA Interference , RNA, Small Interfering/metabolism , Rad51 Recombinase/metabolism
8.
PLoS One ; 8(11): e78961, 2013.
Article in English | MEDLINE | ID: mdl-24244394

ABSTRACT

SLD5 forms a GINS complex with PSF1, PSF2 and PSF3, which is essential for the initiation of DNA replication in lower eukaryotes. Although these components are conserved in mammals, their biological function is unclear. We show here that targeted disruption of SLD5 in mice causes a defect in cell proliferation in the inner cell mass, resulting in embryonic lethality at the peri-implantation stage, indicating that SLD5 is essential for embryogenesis. Moreover, this phenotype of SLD5 mutant mice is quite similar compared with that of PSF1 mutant mice. We have previously reported that haploinsufficiency of PSF1 resulted in failure of acute proliferation of bone marrow hematopoietic stem cells (HSCs) during reconstitution of bone marrow ablated by 5-FU treatment. Since SLD5 was highly expressed in bone marrow, we investigated its involvement in bone marrow reconstitution after bone marrow ablation as observed in PSF1 heterozygous mutant mice. However, heterozygous deletion of the SLD5 gene was found not to significantly affect bone marrow reconstitution. On the other hand, abundant SLD5 expression was observed in human cancer cell lines and heterozygous deletion of the gene attenuated tumor progression in a murine model of spontaneous gastric cancer. These indicated that requirement and dependency of SLD5 for cell proliferation is different in different cell types.


Subject(s)
Carrier Proteins/metabolism , Cell Proliferation , Embryonic Development , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Carrier Proteins/genetics , Female , Gene Deletion , Humans , Male , Mice , Mice, Mutant Strains
9.
Biochem Biophys Res Commun ; 437(4): 609-14, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23867818

ABSTRACT

Fibrosis is one of the most common pathological alterations in heart failure, and fibroblast migration is an essential process in the development of cardiac fibrosis. Experimental autoimmune myocarditis (EAM) is a model of inflammatory heart disease characterized by inflammatory cell infiltration followed by healing without residual fibrosis. However, the precise mechanisms mediating termination of inflammation and nonfibrotic healing remain to be elucidated. Microarray analysis of hearts from model mice at multiple time points after EAM induction identified several secreted proteins upregulated during nonfibrotic healing, including the anti-inflammatory cathelicidin antimicrobial peptide (CAMP). Treatment with LL-37, a human homolog of CAMP, activated MAP kinases in fibroblasts but not in cardiomyocytes, indicating that fibroblasts were the target of CAMP activity. In addition, LL-37 decreased fibroblast migration in the in vitro scratch assay. P2X7 receptor (P2X7R), a well-known receptor for LL-37, was involved in LL-37 mediated biological effect on cardiac fibroblasts. Stimulation of BzATP, a P2X7R agonist, activated MAPK in fibroblasts, whereas the P2X7R antagonist, BBG, as well as P2X7R deletion abolished both LL-37-mediated MAPK activation and LL-37-induced reduction in fibroblast migration. These results strongly suggest that CAMP upregulation during myocarditis prevents myocardial fibrosis by restricting fibroblast migration via activation of the P2X7R-MAPK signaling pathway.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Fibroblasts/drug effects , Receptors, Purinergic P2X7/metabolism , Signal Transduction , Animals , Anti-Inflammatory Agents/pharmacology , Cell Movement , Cells, Cultured , Dose-Response Relationship, Drug , Fibrosis , MAP Kinase Signaling System , Male , Mice , Mice, Inbred BALB C , Phosphorylation , Rats , Cathelicidins
10.
JAKSTAT ; 1(2): 125-30, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-24058761

ABSTRACT

Recently various kinds of cardiac stem/progenitor cells have been identified and suggested to be involved in cardiac repair and regeneration in injured myocardium. In this review, we focus on the roles of JAK-STAT signaling in cardiac stem/progenitor cells in cardiomyogenesis. JAK-STAT signaling plays important roles in the differentiation of stem cells into cardiac lineage cells. The activation of JAK-STAT signal elicits the mobilization of mesenchymal stem cells as well, contributing to the maintenance of cardiac function. Thus we propose that JAK-STAT could be a target signaling pathway in cardiac regenerative therapy.

11.
J Pharmacol Sci ; 117(4): 213-22, 2011.
Article in English | MEDLINE | ID: mdl-22056652

ABSTRACT

Postnatal cardiomyocytes have only limited capacity of proliferation. Therefore, the myocardium is intrinsically equipped with cardioprotective machineries and protects itself from pathological stresses. One of the most important cardioprotective systems is the signal network of autocrine/paracrine factors, including neurohumoral factors, growth factors, and cytokines. In this review, we focus on the roles of interleukin-6 (IL-6) family cytokines, also known as glycoprotein 130 (gp130) cytokines, in cardioprotection. These cytokines make a complex with their specific cytokine receptor α-subunits. The cytokine-receptor α-subunit complex binds to gp130, a common receptor of the IL-6 family, followed by the activation of JAK/STAT, ERK, and PI3 kinase/Akt pathways. In cardiomyocytes, signals through gp130 promote cell survival and angiogenesis through the JAK/STAT pathway. Activation of gp130 in cardiac stem cells induces their endothelial transdifferentiation, leading to neovascularization. Recently, accumulating evidence has revealed that altered JAK/STAT activity is associated with heart failure, suggesting that the JAK/STAT pathway is a therapeutic target against cardiovascular diseases. Interestingly, activation of the JAK/STAT pathway with interleukin-11 (IL-11) exhibits preconditioning effects in ischemia/reperfusion model. Moreover, IL-11 treatment after coronary ligation prevents cardiac remodeling through the JAK/STAT pathway. Since IL-11 is used for patients with thrombocytopenia, we propose that IL-11 is a candidate cytokine clinically available for cardioprotection therapy.


Subject(s)
Cardiovascular Diseases/prevention & control , Cytokine Receptor gp130/metabolism , Drug Delivery Systems , Animals , Cardiotonic Agents/pharmacology , Cardiovascular Diseases/physiopathology , Cytokines/metabolism , Humans , Janus Kinases/metabolism , Myocytes, Cardiac/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/drug effects
12.
J Mol Cell Cardiol ; 51(2): 207-14, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21600215

ABSTRACT

Cardiac stem cells potentially differentiate into cardiac cells, including cardiomyocytes and endothelial cells (ECs). Previously we demonstrated that STAT3 activation by IL-6 family cytokines, such as leukemia inhibitory factor (LIF), induces the endothelial differentiation of cardiac Sca-1+ cells. In this study, we addressed molecular mechanisms for EC differentiation of Sca-1+ cells. First, DNA array experiments were performed to search for the molecules induced by LIF. Among 134 genes that LIF upregulated by more than 4 fold, we focused on Pim-1 gene transcript, because Pim-1 is associated with the differentiation of some cell lineages. Real time RT-PCR analyses confirmed that LIF stimulation upregulated Pim-1 expression. Adenoviral transfection of dominant negative (dn) STAT3 inhibited LIF-mediated induction of Pim-1, while the overexpression of constitutively active STAT3 upregulated Pim-1 expression, suggesting that STAT3 activation is necessary and sufficient for Pim-1 induction. Moreover, in STAT3-deficient Sca-1+ cells, LIF failed to induce Pim-1 expression and EC differentiation. Importantly, the overexpression of dnPim-1 abrogated the induction of EC markers, indicating Pim kinase activity is indispensable for STAT3-mediated EC differentiation in vitro. Finally, Sca-1+ cells labeled with LacZ were transplanted into post-infarct myocardium and the transdifferentiation was estimated. The overexpression of wild-type STAT3 by adenovirus vector significantly promoted EC differentiation, while STAT3 gene ablation reduced the frequency of differentiating cells in post-infarct myocardium. Furthermore, transplanted Sca-1+ cells overexpressing dnPim-1 showed the reduced frequency of EC differentiation and capillary density. Collectively, Pim-1 kinase is upregulated by STAT3 activation in cardiac Sca-1+ cells and plays a pivotal role in EC differentiation both in vitro and in vivo.


Subject(s)
Antigens, Ly/metabolism , Endothelial Cells , Membrane Proteins/metabolism , Myocytes, Cardiac , Proto-Oncogene Proteins c-pim-1/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Stem Cells/metabolism , Animals , Cell Transdifferentiation/genetics , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Leukemia Inhibitory Factor/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Stem Cell Transplantation , Stem Cells/drug effects
13.
Circulation ; 121(5): 684-91, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20100971

ABSTRACT

BACKGROUND: Glycoprotein 130 is the common receptor subunit for the interleukin (IL)-6 cytokine family. Previously, we reported that pretreatment of IL-11, an IL-6 family cytokine, activates the glycoprotein 130 signaling pathway in cardiomyocytes and prevents ischemia/reperfusion injury in vivo; however, its long-term effects on cardiac remodeling after myocardial infarction (MI) remain to be elucidated. METHODS AND RESULTS: MI was generated by ligating the left coronary artery in C57BL/6 mice. Real-time reverse transcription polymerase chain reaction analyses showed that IL-11 mRNA was remarkably upregulated in the hearts exposed to MI. Intravenous injection of IL-11 activated signal transducer and activator of transcription 3 (STAT3), a downstream signaling molecule of glycoprotein 130, in cardiomyocytes in vivo, suggesting that cardiac myocytes are target cells of IL-11 in the hearts. Twenty-four hours after coronary ligation, IL-11 was administered intravenously, followed by consecutive administration every 24 hours for 4 days. IL-11 treatment reduced fibrosis area 14 days after MI, attenuating cardiac dysfunction. Consistent with a previous report that STAT3 exhibits antiapoptotic and angiogenic activity in the heart, IL-11 treatment prevented apoptotic cell death of the bordering myocardium adjacent to the infarct zone and increased capillary density at the border zone. Importantly, cardiac-specific ablation of STAT3 abrogated IL-11-mediated attenuation of fibrosis and was associated with left ventricular enlargement. Moreover, with the use of cardiac-specific transgenic mice expressing constitutively active STAT3, cardiac STAT3 activation was shown to be sufficient to prevent adverse cardiac remodeling. CONCLUSIONS: IL-11 attenuated cardiac fibrosis after MI through STAT3. Activation of the IL-11/glycoprotein 130/STAT3 axis may be a novel therapeutic strategy against cardiovascular diseases.


Subject(s)
Cytokine Receptor gp130/metabolism , Interleukin-11/pharmacology , Myocardial Infarction/drug therapy , STAT3 Transcription Factor/metabolism , Animals , Apoptosis/drug effects , Fibrosis , Gene Expression Regulation/drug effects , Interleukin-11/biosynthesis , Interleukin-6/metabolism , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Neovascularization, Physiologic/drug effects , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
14.
Arterioscler Thromb Vasc Biol ; 29(5): 754-60, 2009 May.
Article in English | MEDLINE | ID: mdl-19213943

ABSTRACT

OBJECTIVE: Cardiac Sca-1+ cells were originally identified as multipotent stem cells. To address the regulation of their differentiation, we investigated the effects of the proinflammatory cytokines on their endothelial differentiation. METHODS AND RESULTS: We examined the effects of the proinflammatory cytokines including tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-1beta, IL-6, IL-11, and cardiotrophin-1 (CT-1) on the cardiac Sca-1+ cell differentiation. IL-11 and CT-1, whose receptor systems use glycoprotein 130 (gp130), induced endothelial-specific genes in Sca-1+ cells, but not TNF-alpha, IL-1beta, or IL-6, analyzed by RT-PCR and by immunocytochemistry. Immnunoblot analyses showed that IL-11 and CT-1 activated signal transducer and activator of transcription 3 (STAT3), a downstream target of gp130, but not other cytokines. Though IL-6 receptor is not endogenously expressed in Sca-1+ cells, IL-6 exhibited the activity to induce the endothelial markers in the presence of soluble IL-6 receptor, an agonistic receptor, associated with STAT3 phosphorylation. Moreover, the inhibition of STAT3, by its dominant-negative form or siRNA, suppressed the induction of endothelial specific genes by IL-11 and CT-1. Finally, LIF and IL-11 transcripts were upregulated in postinfarct myocardium, accompanied by the induction of Sca-1+/VE-cadherin+ cells. CONCLUSIONS: Gp130/STAT3 pathway plays critical roles in the regulation of endothelial differentiation of cardiac Sca-1+ cells.


Subject(s)
Cell Differentiation/physiology , Cytokine Receptor gp130/physiology , Endothelial Cells/physiology , Myocardium/cytology , Signal Transduction , Stem Cells/physiology , Animals , Ataxin-1 , Ataxins , Cells, Cultured , Interleukin-6 , Mice , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Receptors, Interleukin-6 , STAT3 Transcription Factor/physiology
15.
Biochem Biophys Res Commun ; 370(2): 274-8, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18375200

ABSTRACT

In the process of cardiac remodeling, connective tissue growth factor (CTGF/CCN2) is secreted from cardiac myocytes. Though CTGF is well known to promote fibroblast proliferation, its pathophysiological effects in cardiac myocytes remain to be elucidated. In this study, we examined the biological effects of CTGF in rat neonatal cardiomyocytes. Cardiac myocytes stimulated with full length CTGF and its C-terminal region peptide showed the increase in cell surface area. Similar to hypertrophic ligands for G-protein coupled receptors, such as endothelin-1, CTGF activated amino acid uptake; however, CTGF-induced hypertrophy is not associated with the increased expression of skeletal actin or BNP, analyzed by Northern-blotting. CTGF treatment activated ERK1/2, p38 MAPK, JNK and Akt. The inhibition of Akt by transducing dominant-negative Akt abrogated CTGF-mediated increase in cell size, while the inhibition of MAP kinases did not affect the cardiac hypertrophy. These findings indicate that CTGF is a novel hypertrophic factor in cardiac myocytes.


Subject(s)
Cardiomegaly/metabolism , Immediate-Early Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Amino Acids/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Size/drug effects , Cells, Cultured , Connective Tissue Growth Factor , Gene Expression/drug effects , Immediate-Early Proteins/genetics , Immediate-Early Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction
16.
World J Gastroenterol ; 14(16): 2609-11, 2008 Apr 28.
Article in English | MEDLINE | ID: mdl-18442217

ABSTRACT

Gastrointestinal stromal tumor (GIST) of the stomach was treated by endoscopic enucleation in five patients. They were three men and two woman, aged 36-56 years. Tumors located in the cardia were completely enucleated endoscopically without any serious complication. The largest diameter of removed tumors ranged from 1.2 to 2.5 cm. Histopathological diagnosis was GIST with low risk of malignancy (mitotic index < 5/50 high power field) in all cases. The patients were disease-free for 10.5-42.2 mo after endoscopic enucleation.


Subject(s)
Gastrointestinal Stromal Tumors/surgery , Adult , Disease-Free Survival , Endoscopy/methods , Female , Gastrointestinal Stromal Tumors/diagnostic imaging , Gastrointestinal Stromal Tumors/pathology , Humans , Intestinal Mucosa/diagnostic imaging , Intestinal Mucosa/pathology , Male , Middle Aged , Mitotic Index , Ultrasonography
17.
Cardiovasc Res ; 79(1): 89-96, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18346979

ABSTRACT

AIMS: Doxorubicin (DOX) is one of the most effective anti-neoplastic agents; however, its clinical use is limited by drug-induced cardiomyopathy. The molecular mechanisms responsible for this toxicity remain to be fully addressed. In the present study, we investigated the involvement of atrogin-1, one of the muscle-specific ubiquitin ligases, in DOX-induced cardiotoxicity. METHODS AND RESULTS: This method involved intraperitoneal administration of DOX-induced atrogin-1 in the hearts and skeletal muscles of C57BL/6 mice. Consistently, atrogin-1 mRNA was upregulated with DOX treatment in cultured rat neonatal cardiomyocytes. Adenoviral transfer of atrogin-1 induced a reduction in cell size that was ameliorated by the ubiquitin proteasome inhibitor, MG-132. The transduction of constitutively active Akt (caAkt), a serine/threonine protein kinase, inhibited the DOX-mediated induction of atrogin-1. The phosphorylation status of Akt and its downstream target, FOXO, was not affected by DOX. DOX treatment did not activate the atrogin-1 promoter that contains FOXO-binding sites, suggesting that DOX induced atrogin-1 without modulating the Akt/FOXO pathway; importantly, DOX activated p38-mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK). Furthermore, pharmacological inhibition of p38-MAPK, but not JNK, abrogated DOX-mediated induction of atrogin-1. Finally, adenoviral transfer of caAkt inhibited the DOX-induced p38-MAPK activation. CONCLUSIONS: DOX induces atrogin-1 through a p38-MAPK-dependent pathway in cardiac myocytes. Constitutive activation of Akt negatively regulates DOX-mediated atrogin-1 induction by inhibiting p38-MAPK activity as a novel mechanism.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Muscle Proteins/metabolism , Myocytes, Cardiac/enzymology , SKP Cullin F-Box Protein Ligases/metabolism , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Adenoviridae/genetics , Animals , Antibiotics, Antineoplastic/adverse effects , Atrophy , Cardiomyopathies/chemically induced , Cells, Cultured , Doxorubicin/adverse effects , Gene Transfer Techniques , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/drug effects , Muscle Proteins/genetics , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , SKP Cullin F-Box Protein Ligases/drug effects , SKP Cullin F-Box Protein Ligases/genetics , Signal Transduction/drug effects
18.
Am Surg ; 73(9): 895-6, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17939421

ABSTRACT

Peritoneal loose bodies are usually small, 0.5 to 2.5 cm in diameter, and rarely cause symptoms. However, "giant" peritoneal loose bodies, larger than 5 cm in diameter, presenting with various symptoms have been also reported. We report here a case of a "giant" peritoneal loose body found in a 73-year-old man. In this case, the peritoneal loose body was 9.5 cm in diameter, comparable to the largest one reported in the literature. The patient refused any treatment at his first visit but accepted surgical removal of the peritoneal loose body 5 years later. The size of the peritoneal loose body has increased from 7.3 cm to 9.5 cm in diameter during this observation period.


Subject(s)
Calcinosis/diagnostic imaging , Peritoneal Diseases/diagnostic imaging , Abdominal Pain/diagnostic imaging , Aged , Calcinosis/surgery , Diagnosis, Differential , Humans , Male , Peritoneal Diseases/surgery , Tomography, X-Ray Computed
19.
Cytokine ; 38(2): 107-15, 2007 May.
Article in English | MEDLINE | ID: mdl-17629706

ABSTRACT

Interleukin (IL)-6 family cytokines, which share glycoprotein 130 (gp130) as a signal-transducing receptor component, play important roles in the maintenance of cardiac homeostasis. IL-11, a member of IL-6 family cytokines, is expressed in cardiac myocytes, though it remains to be elucidated how IL-11 functions in the hearts. In the present study, first, we showed that IL-11 administration reduced the ischemia/reperfusion injury in the hearts. IL-11 receptor alpha was expressed in cardiomyocytes. IL-11 treatment rapidly activated signal transducer and activator of transcription 3 (STAT3) and extracellular signal-regulated kinase (ERK) 1/2 in cardiac myocytes. IL-11 stimulation resulted in the translocation of phosphorylated STAT3 into nuclei. Immunofluorescence microscopic analyses revealed that IL-11 treatment led to the cell elongation, as is the case with other cardiotrophic members of IL-6 family, such as leukemia inhibitory factor. Finally we showed that IL-11 treatment conferred the resistance to cell death induced by hydrogen peroxide, which was abrogated by adenoviral transfer of dominant negative STAT3, but not by the inhibition of ERK1/2 with U0126. These findings indicate that IL-11 mediates cytoprotective signals in cardiomyocytes, proposing that IL-11 has the potential to exhibit cardioprotection as a novel biological function.


Subject(s)
Cardiotonic Agents/metabolism , Cardiotonic Agents/therapeutic use , Interleukin-11/physiology , Myocytes, Cardiac/metabolism , Animals , Humans , Interleukin-11/therapeutic use , Mice , Mice, Inbred C57BL , Reperfusion Injury/therapy , STAT3 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...