Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 50
1.
PLoS One ; 19(5): e0303758, 2024.
Article En | MEDLINE | ID: mdl-38768136

Nitric oxide (NO) promotes angiogenesis via various mechanisms; however, the effective transmission of NO in ischemic diseases is unclear. Herein, we tested whether NO-releasing nanofibers modulate therapeutic angiogenesis in an animal hindlimb ischemia model. Male wild-type C57BL/6 mice with surgically-induced hindlimb ischemia were treated with NO-releasing 3-methylaminopropyltrimethoxysilane (MAP3)-derived or control (i.e., non-NO-releasing) nanofibers, by applying them to the wound for 20 min, three times every two days. The amount of NO from the nanofiber into tissues was assessed by NO fluorometric assay. The activity of cGMP-dependent protein kinase (PKG) was determined by western blot analysis. Perfusion ratios were measured 2, 4, and 14 days after inducing ischemia using laser doppler imaging. On day 4, Immunohistochemistry (IHC) with F4/80 and gelatin zymography were performed. IHC with CD31 was performed on day 14. To determine the angiogenic potential of NO-releasing nanofibers, aorta-ring explants were treated with MAP3 or control fiber for 20 min, and the sprout lengths were examined after 6 days. As per either LDPI (Laser doppler perfusion image) ratio or CD31 capillary density measurement, angiogenesis in the ischemic hindlimb was improved in the MAP3 nanofiber group; further, the total nitrate/nitrite concentration in the adduct muscle increased. The number of macrophage infiltrations and matrix metalloproteinase-9 (MMP-9) activity decreased. Vasodilator-stimulated phosphoprotein (VASP), one of the major substrates for PKG, increased phosphorylation in the MAP3 group. MAP3 nanofiber or NO donor SNAP (s-nitroso-n-acetyl penicillamine)-treated aortic explants showed enhanced sprouting in an ex vivo aortic ring assay, which was partially abrogated by KT5823, a potent inhibitor of PKG. These findings suggest that the novel NO-releasing nanofiber, MAP3 activates PKG and promotes therapeutic angiogenesis in response to hindlimb ischemia.


Cyclic GMP-Dependent Protein Kinases , Hindlimb , Ischemia , Mice, Inbred C57BL , Nanofibers , Neovascularization, Physiologic , Nitric Oxide , Animals , Nanofibers/chemistry , Male , Nitric Oxide/metabolism , Ischemia/drug therapy , Ischemia/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Mice , Hindlimb/blood supply , Neovascularization, Physiologic/drug effects , Matrix Metalloproteinase 9/metabolism , Phosphoproteins/metabolism , Microfilament Proteins/metabolism , Cell Adhesion Molecules
2.
J Biochem Mol Toxicol ; 37(3): e23267, 2023 Mar.
Article En | MEDLINE | ID: mdl-36524533

We identified a gene, subunit C3 (ATP5G3) of mitochondrial ATP synthase, that displayed changes in gene expression under oxidative stress. We examined the role of ATP5G3 and its molecular mechanisms in sodium nitroprusside (SNP)-induced cell death using ATP5G3 small interfering RNA (siATP5G3)-transfected HeLa cells. A significant increase in cytotoxicity was observed in the transfected cells treated with SNP, which suggests a protective role of ATP5G3 in SNP-induced cytotoxicity in the cells. The transfected cells treated with photodegraded SNP showed equal cytotoxicity to SNP, and pretreatment with deferoxamine (DFO) completely inhibited this cytotoxicity. Further, cytotoxicity was significantly inhibited by pretreatment with a p38 inhibitor and was accentuated by the p38 activator in cells. Pretreatment with the Bcl-xL inhibitor also significantly accentuated cytotoxicity. The increase in p38 phosphorylation was significantly higher in siATP5G3-transfected cells treated with SNP in immunoblotting, which was inhibited by pretreatment with DFO. The increase in cytotoxicity with siATP5G3 transfection was completely blocked by cotransfection with sip38, and the blocking effect disappeared by cotransfection with additional siBcl-xL, which suggests that the protective role of ATP5G3 is mediated by Bcl-xL via the inhibition of p38 activity. Cytotoxicity was completely blocked by the cotransfection of siATP5G3 with siBax. No change in apoptotic parameters was observed during cytotoxicity. However, pretreatment with lysosomal inhibitors significantly inhibited cytotoxicity and increased p62 protein levels. These findings suggest that ATP5G3 plays a protective role in autophagic cell death/lysosome-associated cell death induced by SNP via the sequential signaling of ROS/p38/Bcl-xL/Bax in HeLa cells.


Carcinoma , Humans , Apoptosis , Cell Death , Cell Line, Tumor , HeLa Cells , Nitroprusside/pharmacology
3.
Life (Basel) ; 12(2)2022 Jan 28.
Article En | MEDLINE | ID: mdl-35207485

Dyslipidemia, the commonest cause of cardiovascular disease, leads to lipid deposits on the arterial wall, thereby aggravating atherosclerosis. DSHT (Daeshiho-tang) has long been used as an anti-dyslipidemia agent in oriental medicine. However, the anti-atherosclerotic effects of DSHT have not been fully investigated. Therefore, this study was designed to evaluate whether DSHT could exert beneficial anti-atherosclerotic effects. We fed apolipoprotein E-deficient (ApoE-/-) mice on a high-fat diet and treated them with atorvastatin (AT) or DSHT, or the combination of DSHT and AT for 12 weeks. To determine the role of DSHT, atherosclerotic lesions in the aorta, aortic root, and aortic arch; lipids and apolipoprotein levels in serum; and macrophage polarization markers in aorta tissues were examined. We show here that the DSHT decreased the atherosclerotic plaque ratio in the aortic arch, aorta, and aortic root. DSHT also regulated lipid levels by decreasing the ApoB level and increasing the ApoA1 level. Moreover, DSHT effectively regulated cholesterol metabolism by increasing the levels of PPARγ, ABCA1 and ABCG1, and the LDL receptor genes. We further found that DSHT promoted polarization to the M2 phenotype by increasing the levels of M2 macrophage (ARG1, CD163, and PPARγ) markers. Our data suggested that DSHT enhances the anti-atherosclerotic effect by regulating cholesterol metabolism through the activation of the PPARγ signaling pathway and by promoting anti-inflammatory M2 macrophage polarization.

4.
Am J Cardiovasc Drugs ; 22(2): 117-125, 2022 Mar.
Article En | MEDLINE | ID: mdl-34151411

The anti-cancer agent doxorubicin (DOX) has high cardiotoxicity that is linked to DOX-mediated increase in oxidative stress, mitochondrial iron overload, DNA damage, autophagy, necrosis, and apoptosis, all of which are also associated with secondary tumorigenicity. This limits the clinical application of DOX therapies. Previous studies have attributed DOX-mediated cardiotoxicity to mitochondrial iron accumulation and the production of reactive oxygen species (ROS), which seem to be independent of its anti-tumor DNA damaging effects. Chemo-sensitization of soluble guanylate cyclase (sGC) in the cyclic guanosine monophosphate (cGMP) pathway induces tumor cell death despite the cardiotoxicity associated with DOX treatment. However, sGC-cGMP signaling must be activated during heart failure to facilitate myocardial cell survival. The sGC pathway is dependent on nitric oxide and signal transduction via the nitric oxide-sGC-cGMP pathway and is attenuated in various cardiovascular diseases. Additionally, cGMP signaling is regulated by the action of certain phosphodiesterases (PDEs) that protect the heart by inhibiting PDE, an enzyme that hydrolyses cGMP to GMP activity. In this review, we discuss the studies describing the interactions between cGMP regulation and DOX-mediated cardiotoxicity and their application in improving DOX therapeutic outcomes. The results provide novel avenues for the reduction of DOX-induced secondary tumorigenicity and improve cellular autonomy during DOX-mediated cardiotoxicity.


Cyclic GMP , Heart Failure , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Doxorubicin/adverse effects , Heart Failure/chemically induced , Heart Failure/drug therapy , Humans , Signal Transduction , Soluble Guanylyl Cyclase/metabolism , Soluble Guanylyl Cyclase/pharmacology
5.
Chem Biol Interact ; 328: 109190, 2020 Sep 01.
Article En | MEDLINE | ID: mdl-32652078

BACKGROUND: Doxorubicin (DOX) administration decreases cardiac soluble guanylate cyclase (sGC) activity. We hypothesized that bypassing impaired NO-sGC-cGMP pathway resulting from the activation of oxidized and heme-free soluble guanylate cyclase (sGC) could be a therapeutic target for DOX-mediated cardiomyopathy (DOX-CM). The present study investigated the therapeutic roles and mechanism of BAY60-2770, an activator of oxidized sGC, in alleviating DOX-CM. METHODS: H9c2 cardiomyocytes were pretreated with BAY60-2770 followed by DOX. Cell viability and intracellular reactive oxygen species (ROS) were subsequently measured. To determine the role BAY60-2770 in mitochondrial ROS generation and mitochondrial membrane potential, we examined mitoSOX RED and TMRE fluorescence under DOX exposure. As animal experiments, rats were orally administered with 5 mg/kg of BAY60-2770 at 1 h prior to every DOX treatment and then assessed by echocardiography and apoptotic marker and autophagy. RESULTS: BAY60-2770 ameliorated cell viability and DOX-induced oxidative stress in H9c2 cells, which was mediated by PKG activation. Mitochondrial ROS and TMRE fluorescence were attenuated by BAY60-2770 in DOX-treated H9c2 cells. DOX-induced caspase-3 activation decreased after pretreatment with BAY60-2770 in vivo and in vitro. Echocardiography showed that BAY60-2770 significantly improved DOX-induced myocardial dysfunction. Autophagosome was increased by BAY60-2770 in vivo. CONCLUSIONS: BAY60-2770 appears to mitigate DOX-induced mitochondrial ROS, membrane potential loss, autophagy, and subsequent apoptosis, leading to protection of myocardial injury and dysfunction. These novel results highlighted the therapeutic potential of BAY60-2770 in preventing DOX-CM.


Autophagy/drug effects , Benzoates/pharmacology , Biphenyl Compounds/pharmacology , Cardiotoxicity/pathology , Doxorubicin/adverse effects , Hydrocarbons, Fluorinated/pharmacology , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Cell Line , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
6.
BMC Mol Cell Biol ; 21(1): 41, 2020 Jun 09.
Article En | MEDLINE | ID: mdl-32517655

BACKGROUND: Localization of neurokinin 1 receptor (NK1R), the endogenous receptor for neuropeptide substance P (SP), has already been described for the right atrium (RA) of the heart. However, the biological role of SP/NK1R signal pathways in the RA remains unclear. Sprague-Dawley rats were randomly divided into 4 groups (n = 22 each); subjected to sham, ischemia/reperfusion-injury (I/R), I/R with 5 nmole/kg SP injection (SP + I/R), and SP + I/R with 1 mg/kg RP67580 injection (RP, a selective non-peptide tachykinin NK1R antagonist) (RP/SP + I/R). The left anterior descending coronary artery was occluded for 40 min followed by 1 day reperfusion with SP or SP + RP or without either. After 1 day, both atria and ventricles as well as the heart apexes were collected. RESULTS: SP promoted the expression of c-Kit, GATA4, Oct4, Nanog, and Sox2 in only the RA of the SP + I/R rats via NK1R activation. In agreement with these observations, NK1R-expressing c-Kit+ Nkx2.5+GATA4+ cardiac progenitor cells (CPCs) in the ex vivo RA explant outgrowth assay markedly migrated out from RA1 day SP + I/R approximately 2-fold increase more than RA1 day I/R. Treatment of SP promoted proliferation, migration, cardiosphere formation, and potential to differentiate into cardiomyocytes. Using RP inhibitor, NK1R antagonist not only inhibited cell proliferation and migration but also reduced the formation of cardiosphere and differentiation of c-Kit+ CPCs. CONCLUSION: SP/NK1R might play a role as a key mediator involved in the cellular response to c-Kit+ CPC expansion in RA of the heart within 24 h after I/R.


Heart Atria/metabolism , Multipotent Stem Cells/metabolism , Reperfusion Injury , Substance P/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Heart Injuries , Neurokinin-1 Receptor Antagonists/pharmacology , Proto-Oncogene Proteins c-kit/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/metabolism , Signal Transduction/drug effects
7.
Cell Physiol Biochem ; 52(1): 40-56, 2019.
Article En | MEDLINE | ID: mdl-30790504

BACKGROUND/AIMS: Therapies using stem/progenitor cells have been experimentally and clinically investigated to regenerate damaged hearts. Substance-P (SP) induces bone marrow (BM) stem cell mobilization and suppresses inflammation in ischemic injuries. This study investigated the role of SP in BM stem cell mobilization and immune responses for tissue repair after ischemic-reperfusion injury (IRI), in comparison with that of granulocyte colony-stimulating factor (GCSF). METHODS: SP was intravenously injected into IRI rats and its affect was evaluated by determining colony forming efficiency, immune cell/ cytokine profiles, histological changes, and heart function through echocardiography. RESULTS: In the rat cardiac IRI model, SP suppressed IRI-mediated tumor necrosis factor-α induction, but increased the levels of interleukin-10, CD206+ monocytes, and regulatory T cells in the blood; reduced myocardial apoptosis at day 1 post-IRI; and markedly stimulated colony forming unit (CFU)-e and (CFU)-f cell mobilization. Efficacy of SP in the recovery of cardiac function after IRI was demonstrated by increased cardiac contractility, accompanied by reduced infarction sizes and fibrosis, and increased revascularization of vessels covered with alpha smooth muscle actin. These effects of SP were confirmed in an acute myocardial infarction (AMI) model. All effects mediated by SP were superior to those mediated by GCSF. CONCLUSION: Systemic injection of SP decreased early inflammatory responses and promoted stem cell mobilization, leading to a compact vasculature and improved cardiac function in cardiac IRI and AMI.


Hematopoietic Stem Cell Mobilization , Myocardial Infarction , Myocardial Reperfusion Injury , Substance P/pharmacokinetics , Animals , Granulocyte Colony-Stimulating Factor/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Interleukin-10/metabolism , Lectins, C-Type/metabolism , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Exp Mol Med ; 50(8): 1-15, 2018 08 15.
Article En | MEDLINE | ID: mdl-30111834

The peroxisome proliferator-activated receptor-γ (PPARγ) improves whole-body insulin sensitivity by regulating the adipogenic and metabolic functions of mature adipocytes. We have previously demonstrated that an active splice variant of X-box binding protein 1 (XBP1s) enhances PPARγ expression during adipogenesis. In this study, we investigated the role of XBP1s, particularly with respect to PPARγ, in the mechanisms underlying insulin sensitivity in mature adipocytes. Insulin was able to stimulate XBP1s generation by activating inositol-requiring enzyme 1 (IRE1) α and was also able to increase its transcriptional activity by inducing nuclear translocation. XBP1s also upregulated the levels of phosphorylated IRS1 and AKT, demonstrating a positive feedback regulatory mechanism linking insulin and XBP1s. XBP1s enhanced the expression of fibroblast growth factor 21 and, in turn, increased PPARγ activity, translocation of GLUT4 to the cell surface, and glucose uptake rate in adipocytes. In addition, XBP1s abolished palmitate-induced insulin resistance in adipocytes by increasing adiponectin secretion, repressing the secretion of pro-inflammatory adipokines such as leptin, monocyte chemoattractant protein 1, and tumor necrosis factor α, and decreasing fatty acid release. These findings provide a novel mechanism by which XBP1s stimulate insulin sensitivity in adipocytes through fibroblast growth factor 21 induction and PPARγ activation.


Adipocytes/metabolism , Endoribonucleases/metabolism , Glucose/metabolism , Insulin/pharmacology , PPAR gamma/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , X-Box Binding Protein 1/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Fibroblast Growth Factors/metabolism , Insulin Resistance , Mice , Models, Biological , Palmitic Acid , Protein Transport/drug effects , RNA Splicing/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , X-Box Binding Protein 1/genetics
9.
Int J Cardiol ; 271: 228-232, 2018 Nov 15.
Article En | MEDLINE | ID: mdl-29884290

BACKGROUND: Substance P (SP) may attenuate ischemia-reperfusion injury by reducing inflammation. We assessed cardioprotective effect of SP in a porcine model of acute myocardial infarction (AMI). METHODS: AMI was induced by occlusion of the left anterior descending artery on 28 swine, randomized to SP 5 nmol/kg (group 1, n = 14) and normal saline (group 2, n = 14) given intravenously 5 min before reperfusion. Blood samples were collected at baseline, 3 days and 4 weeks. Echocardiography and myocardial perfusion single photon emission computed tomography (SPECT) were performed at 1 week and 4 weeks. Histomorphometric infarct size assessment was done at 4 weeks. RESULTS: Left ventricular (LV) ejection fraction (EF) (LVEF) after AMI induction was higher in group 1 than group 2 (37.9 ±â€¯4.6% vs. 29.4 ±â€¯3.2%, p = 0.001) but not different at 4 weeks. No significant difference was observed in perfusion defect extent and total perfusion defect on SPECT at 1 week and 4 weeks. Pathologic infarct size (% LV) was significantly smaller in group 1 than group 2 (2.4 ±â€¯2.3% vs. 5.7 ±â€¯2.5%, p = 0.020). The ratio of neutrophil to lymphocyte on day 3 and serum creatinine concentration at 4 weeks after AMI were lower in group 1. CONCLUSIONS: In a porcine model of AMI, SP improved LVEF early post-MI and reduced infarct size. SP may be beneficial in reducing inflammation and ischemia-reperfusion injury after AMI.


Cardiotonic Agents/administration & dosage , Disease Models, Animal , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/prevention & control , Substance P/administration & dosage , Animals , Infusions, Intravenous , Male , Myocardial Reperfusion/methods , Random Allocation , Swine , Treatment Outcome
10.
Sci Rep ; 7(1): 13718, 2017 10 20.
Article En | MEDLINE | ID: mdl-29057951

Far-infrared radiation (FIR) has been shown to exert positive effects on the cardiovascular system. However, the biological effects of FIR on bone marrow-derived stem cells (BMSCs) are not understood. In the present study, BMSCs were isolated from rat femur bone marrow and cultured in vitro. To investigate the effects of an FIR generator with an energy flux of 0.13 mW/cm2 on rat BMSCs, survival of BMSCs was measured by crystal violet staining, and cell proliferation was additionally measured using Ez-Cytox cell viability, EdU, and Brd U assays. FIR preconditioning was found to significantly increase BMSC proliferation and survival against H2O2. The scratch and transwell migration assays showed that FIR preconditioning resulted in an increase in BMSC migration. qRT-PCR and Western blot analyses demonstrated that FIR upregulated Nanog, Sox2, c-Kit, Nkx2.5, and CXCR4 at both the mRNA and protein levels. Consistent with these observations, PD98059 (an ERK inhibitor) and AMD3100 (a CXCR4 inhibitor) prevented the activation of CXCR4/ERK and blocked the cell proliferation and migration induced by FIR. Overall, these findings provide the first evidence that FIR confers a real and significant benefit on the preconditioning of BMSCs, and might lead to novel strategies for improving BMSC therapy for cardiac ischemia.


Bone Marrow Cells/radiation effects , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Infrared Rays , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Bone Marrow Cells/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Radiation , Extracellular Signal-Regulated MAP Kinases/metabolism , Femur , Gene Expression Regulation/radiation effects , Hydrogen Peroxide/administration & dosage , Hydrogen Peroxide/metabolism , MAP Kinase Signaling System/radiation effects , Male , Rats, Sprague-Dawley , Receptors, CXCR4/metabolism , Time Factors
11.
PLoS One ; 12(7): e0180207, 2017.
Article En | MEDLINE | ID: mdl-28671970

Soluble guanylate cyclase (sGC) has been suggested as a therapeutic target for cardiac ischemia-reperfusion (IR) injury. Until now, the molecular mechanism of BAY 60-2770, a sGC activator, in cardiac IR injury has not been assessed. To identify the cardioprotective effects of BAY 60-2770 in IR-injured rat hearts, IR injury was established by occlusion of LAD for 40 min and reperfusion for 7 days, and the effects of BAY 60-2770 on myocardial protection were assessed by echocardiography and TTC staining. 5 nM and 5 µM of BAY 60-2770 were perfused into isolated rat hearts in a Langendorff system. After 10- or 30-min reperfusion with BAY 60-2770, cGMP and cAMP concentrations and PKG activation status were examined. Hearts were also perfused with 1 µM KT5823 or 100 µM 5-HD in conjunction with 5 nM Bay 60-2770 to evaluate the protective role of PKG. Mitochondrial oxidative stress was investigated under hypoxia-reoxygenation in H9c2 cells. In IR-injured rat hearts, BAY 60-2770 oral administration reduced infarct size by TTC staining and improved left ventricular function by echocardiography. Tissue samples from BAY 60-2770-perfused hearts had approximately two-fold higher cGMP levels. BAY 60-2770 increased PKG activity in the myocardium, and the reduced infarct area by BAY 60-2770 was abrogated by KT-5823 in isolated myocardium. In H9c2 cardiac myoblasts, hypoxia-reoxygenation-mediated mitochondrial ROS generation was diminished with BAY 60-2770 treatment, but was recovered by pretreatment with KT-5823. BAY 60-2770 demonstrated a protective effect against cardiac IR injury via mitoKATP opening and decreased mitoROS by PKG activation. BAY 60-2770 has a protective effect against cardiac IR injury via mitoKATP opening and decreased mitoROS by PKG activation. These results demonstrated that BAY 60-2770 may be used as a therapeutic agent for cardiac IR injury.


Benzoates/pharmacology , Biphenyl Compounds/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Guanylate Cyclase/metabolism , Hydrocarbons, Fluorinated/pharmacology , Myocardial Reperfusion Injury , Animals , Cell Line , Enzyme Activation , In Vitro Techniques , Male , Mitochondria, Heart/metabolism , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Superoxides/metabolism
12.
Biochem Biophys Res Commun ; 488(3): 477-482, 2017 07 01.
Article En | MEDLINE | ID: mdl-28522296

In obese patients, free fatty acids ectopically accumulated in non-adipose tissues cause cell death. Saturated fatty acids are more deleterious to non-adipose cells, and supplementation with monounsaturated fatty acids has been proposed to rescue cells from saturated fatty acid-induced cytotoxicity; however, the mechanisms are not well understood. To understand the cytoprotective role of monounsaturated fatty acids in lipotoxic cell death of macrophages, we investigated the antagonizing effect of oleate and the underlying mechanisms in palmitate-treated RAW264.7 cells. Palmitate strongly induced apoptosis in macrophages by increasing CD36 expression, which was identified to mediate both endoplasmic reticulum stress and the generation of reactive oxygen species. Co-treatment with oleate significantly reduced CD36 expression and its downstream signaling pathways of apoptosis in palmitate-treated cells. These findings provide a novel mechanism by which oleate protects macrophages from palmitate-induced lipotoxicity.


Apoptosis/drug effects , CD36 Antigens/biosynthesis , Down-Regulation/drug effects , Macrophages/drug effects , Oleic Acid/pharmacology , Palmitates/pharmacology , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Macrophages/metabolism , Mice , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
13.
Int J Cardiol ; 232: 40-47, 2017 Apr 01.
Article En | MEDLINE | ID: mdl-28159361

OBJECTIVES: Exenatide is a glucagon-like peptide-1 analogue that mitigates myocardial injury caused by ischemia-reperfusion injury via the survival signaling pathway. We hypothesized that exenatide would provide a protective effect in doxorubicin-induced cardiotoxicity. METHODS: H9c2 cardiomyocytes were pre-treated with exenatide followed by doxorubicin (DOX), and cell viability and intracellular reactive oxygen species (ROS) were subsequently measured. In order to determine the role of autophagy, we performed western blot as well as TUNEL and autophagosome staining. Additionally, rats were treated with exenatide 1h prior to every DOX treatment. Left ventricular (LV) function and performance were then assessed by echocardiography. Myocardial and serum ROS was measured with DHE fluorescence and ROS/RNS assay. RESULTS: DOX-induced caspase-3 activation decreased after pre-treatment with exenatide both in vivo and in vitro. Oxidative stress was attenuated by exenatide in H9c2 cells, as well as in cardiac tissue and serum. The number of autophagosomes and autophagic markers were further increased by exenatide in the DOX-treated H9c2 cells, which mediated AMPK activation. Suppression of the autophagosome abolished exenatide-induced anti-apoptotic effect. Echocardiography showed that pre-treatment with exenatide significantly improved LV dysfunction that is induced by DOX treatment. Exenatide inhibits the DOX-induced production of intracellular ROS and apoptosis in the myocardium. The autophagic markers increased in exenatide pre-treated cardiac tissue. CONCLUSION: Exenatide reduces DOX-induced apoptosis of cardiomyocytes by upregulating autophagy and improving cardiac dysfunction. These novel results highlight the therapeutic potential of exenatide to prevent doxorubicin cardiotoxicity.


Apoptosis , Autophagy , Cardiotoxicity/pathology , Heart Ventricles/physiopathology , Myocardium/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , Blotting, Western , Cardiotoxicity/metabolism , Cardiotoxicity/physiopathology , Cell Line , Cell Survival , Disease Models, Animal , Doxorubicin/toxicity , Echocardiography , Heart Ventricles/diagnostic imaging , In Situ Nick-End Labeling , Male , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction , Ventricular Function, Left
14.
Antioxid Redox Signal ; 26(2): 70-83, 2017 01 10.
Article En | MEDLINE | ID: mdl-27357096

AIMS: Doxorubicin (DOX) is a chemotherapeutic drug that is used to treat many cancers, but its use is limited by cardiotoxic side effect. Carbonyl reductase 1 (CBR1) is an NADPH-dependent oxidoreductase that reduces DOX to doxorubicinol (DOXOL), a less potent derivative that is responsible for DOX cardiotoxicity. Thus, we aimed to demonstrate that inhibition of CBR1 enhances the chemotherapeutic efficacy of DOX and attenuates cardiotoxicity. RESULTS: Pharmacological or genetic inhibition of CBR1 improved the anticancer effects of DOX in preclinical models of breast cancer. RNA interference or chemical inhibition of CBR1 improved the anticancer effect of DOX in breast cancer. Moreover, CBR1 overexpression enabled breast cancer cells to obtain chemotherapeutic resistance to DOX treatment. Intriguingly, inhibition of CBR1 decreased DOX-induced cardiotoxicity in animal model. Innovation and Conclusions: Inhibition of CBR1 increases chemotherapeutic efficacy of DOX and reduces cardiotoxicity by blocking DOX reduction to DOXOL. Therefore, we offer preclinical proof-of-concept for a combination strategy to safely leverage the efficacy of doxorubicin by blunting its cardiotoxic effects that limit use of this cytotoxic agent used widely in the oncology clinic. Antioxid. Redox Signal. 26, 70-83.


Alcohol Oxidoreductases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Doxorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers , Breast Neoplasms/drug therapy , Cardiotoxicity , Cell Death/drug effects , Cell Line, Tumor , Creatine Kinase, MB Form/metabolism , Disease Models, Animal , Drug Synergism , Female , Gene Expression , Gene Knockdown Techniques , Humans , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Rats , Xenograft Model Antitumor Assays
15.
Heart Lung Circ ; 26(5): 519-523, 2017 May.
Article En | MEDLINE | ID: mdl-27743854

BACKGROUND: Exenatide exerts cardioprotective effects by attenuating ischaemic reperfusion (IR) injury, possibly through activating the opening of mitochondrial ATP-sensitive potassium channels. We used atomic force microscopy (AFM) to investigate changes in mitochondrial morphology and properties in order to assess exenatide-mediated cardioprotection in IR injury. METHODS: We used an in vivo Sprague-Dawley rat IR model and ex vivo Langendorff injury model. In the left anterior descending artery (LAD) occlusion model, animals were randomly divided into three groups: sham-operated rats (Sham, n=5), IR-injured rats treated with placebo (IR, n=6), and IR-injured treated with exenatide (IR + EXE, n=6). For the Langendorff model, rats were randomly divided into two groups: IR injury with placebo (IR, n=4) and IR injury with exenatide (IR+EXE, n=4). Morphological and mechanical changes of mitochondria were analysed by AFM. RESULTS: Exenatide pre-treatment improved cardiac function as evidenced by improvement in echocardiographic results. The ratio of infarct area (IA) to risk area (RA) was significantly reduced in exenatide-treated rats. According to AFM, IR significantly increased the area of isolated mitochondria, indicative of mitochondrial swelling. Treatment with exenatide reduced the mitochondrial area and ameliorated the adhesion force of mitochondrial surfaces. CONCLUSIONS: Exenatide pre-treatment improves morphological and mechanical characteristics of mitochondria in response to IR injury in a rat model. These alterations in mitochondrial characteristics appear to play a cardioprotective role against IR injury.


Echocardiography , Mitochondria, Heart , Myocardial Reperfusion Injury , Peptides/pharmacology , Venoms/pharmacology , Animals , Disease Models, Animal , Exenatide , Male , Microscopy, Atomic Force , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , Rats , Rats, Sprague-Dawley
16.
Trials ; 17(1): 422, 2016 08 24.
Article En | MEDLINE | ID: mdl-27558002

BACKGROUND: Atrial fibrillation (AF) is known to be associated with several pathophysiological mechanisms including endothelial dysfunction of the heart and arterial vessels. Recent evidence suggests that new oral anticoagulant (NOAC) treatment may improve endothelial function and the inflammatory process involved in atherosclerosis in AF patients. This study is designed to determine the efficacy of NOAC therapy in the prevention of endothelial dysfunction and the progression of atherosclerosis of AF subjects. METHOD/DESIGN: AF patients with a CHA2DS2-VASc score >2 and no previous history of overt coronary disease, severe peripheral arterial disease (PAD) or major stroke will be registered and randomly assigned either to the NOAC group (dabigatran or rivaroxaban) or the warfarin group in this prospective, randomized, 2-year follow-up study. Reactive hyperemia peripheral arterial tonometry (RH-PAT) measurements reflecting endothelial function will be conducted using the Endo-PAT2000 device. Left and right carotid intima-media thickness (IMT) will be measured at baseline, 12 months, and 24 months. The primary endpoint is defined as change in Reactive Hyperemia Index (RHI) at 12 months. Secondary endpoints included changes in the right and left maximum IMT of the common carotid artery (CCA) and internal carotid artery (ICA), the mean IMT of the CCA and ICA at 24 months, and 24-month cardiovascular events including cardiac death, stroke, acute myocardial infarction (AMI), overall cause of death, withdrawal of drug, or bleeding events. DISCUSSION: This is the first study to evaluate the efficacy of NOAC therapy for the prevention of endothelial dysfunction and progression of atherosclerosis in AF subjects. These findings are expected to expand the knowledge of NOAC pleotropic action in AF patients. TRIAL REGISTRATION: ClinicalTrials.gov: NCT02544932 , registered on 7 September 2015.


Anticoagulants/therapeutic use , Atherosclerosis/prevention & control , Atrial Fibrillation/drug therapy , Clinical Protocols , Administration, Oral , Adult , Aged , Aged, 80 and over , Atrial Fibrillation/complications , Humans , Middle Aged , Prospective Studies , Sample Size
20.
PLoS One ; 11(3): e0150745, 2016.
Article En | MEDLINE | ID: mdl-26959365

OBJECTIVE: Although dipeptidyl peptidase-4 (DPP-4) inhibitors have been suggested to have a non-glucoregulatory protective effect in various tissues, the effects of long-term inhibition of DPP-4 on the micro- and macro-vascular complications of type 2 diabetes remain uncertain. The aim of the present study was to investigate the organ-specific protective effects of DPP-4 inhibitor in rodent model of type 2 diabetes. METHODS: Eight-week-old diabetic and obese db/db mice and controls (db/m mice) received vehicle or one of two doses of gemigliptin (0.04 and 0.4%) daily for 12 weeks. Urine albumin excretion and echocardiography measured at 20 weeks of age. Heart and kidney tissue were subjected to molecular analysis and immunohistochemical evaluation. RESULTS: Gemigliptin effectively suppressed plasma DPP-4 activation in db/db mice in a dose-dependent manner. The HbA1c level was normalized in the 0.4% gemigliptin, but not in the 0.04% gemigliptin group. Gemigliptin showed a dose-dependent protective effect on podocytes, anti-apoptotic and anti-oxidant effects in the diabetic kidney. However, the dose-dependent effect of gemigliptin on diabetic cardiomyopathy was ambivalent. The lower dose significantly attenuated left ventricular (LV) dysfunction, apoptosis, and cardiac fibrosis, but the higher dose could not protect the LV dysfunction and cardiac fibrosis. CONCLUSION: Gemigliptin exerted non-glucoregulatory protective effects on both diabetic nephropathy and cardiomyopathy. However, high-level inhibition of DPP-4 was associated with an organ-specific effect on cardiovascular complications in type 2 diabetes.


Cardiovascular Diseases/complications , Cardiovascular Diseases/drug therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Albuminuria/blood , Albuminuria/complications , Animals , Apoptosis/drug effects , Cardiomegaly/blood , Cardiomegaly/complications , Cardiomegaly/physiopathology , Cardiovascular Diseases/blood , Diabetes Mellitus, Type 2/blood , Dipeptidyl Peptidase 4/blood , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Disease Models, Animal , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Glucagon-Like Peptide 1/blood , Glycated Hemoglobin/metabolism , Immunohistochemistry , Kidney/drug effects , Kidney/pathology , Male , Mice , NADPH Oxidases/metabolism , Piperidones/pharmacology , Piperidones/therapeutic use , Podocytes/drug effects , Podocytes/pathology , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Ventricular Dysfunction/blood , Ventricular Dysfunction/complications , Ventricular Dysfunction/physiopathology
...