Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Drug Discov Today ; 27(1): 234-245, 2022 01.
Article in English | MEDLINE | ID: mdl-34438076

ABSTRACT

Undermining new mediators involved in the development and progression of cardiovascular diseases (CVDs) is vital for better disease management. Existing studies implicate a crucial role for inflammation and inflammatory cells, particularly mast cells, in cardiac diseases. Interestingly, the mast cell mediator, histamine, and its receptors profoundly impact the pathophysiology of the heart, resulting in hypertension-induced cardiac hypertrophy and other cardiac anomalies. In this review, we provide a detailed description of mast cell activation, mediators, and histamine receptors, with a particular focus on histamine 2 receptors (H2Rs). Preclinical and clinical studies using histamine receptor antagonists report improvement in cardiac function. Insights into the precise function of histamine receptors will aid in developing novel therapies and pave the way for repurposing antihistamines for cardiovascular diseases.


Subject(s)
Cardiovascular Diseases , Histamine Antagonists/pharmacology , Inflammation , Mast Cells , Receptors, Histamine H2/metabolism , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Histamine/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Mast Cells/drug effects , Mast Cells/physiology
3.
J Cell Biochem ; 121(12): 4887-4897, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32628320

ABSTRACT

Elevated cellular oxidative stress and oxidative DNA damage are key contributors to impaired cardiac function in diabetes. During chronic inflammation, reactive oxygen species (ROS)-induced lipid peroxidation results in the formation of reactive aldehydes, foremost of which is 4-hydroxy-2-nonenal (4HNE). 4HNE forms covalent adducts with proteins, negatively impacting cellular protein function. During conditions of elevated oxidative stress, oxidative DNA damage such as modification by 8-hydroxydeoxyguanosine (8OHdG) is repaired by 8-oxoguanine glycosylase-1 (OGG-1). Based on these facts, we hypothesized that 4HNE forms adducts with OGG-1 inhibiting its activity, and thus, increases the levels of 8OHG in diabetic heart tissues. To test our hypothesis, we evaluated OGG-1 activity, 8OHG and 4HNE in the hearts of leptin receptor deficient db/db mice, a type-2 diabetic model. We also treated the recombinant OGG-1 with 4HNE to measure direct adduction. We found decreased OGG-1 activity (P > .05), increased 8OHG (P > .05) and increased 4HNE adducts (P > .05) along with low aldehyde dehydrogenase-2 activity (P > .05). The increased colocalization of OGG-1 and 4HNE in cardiomyocytes suggest 4HNE adduction on OGG-1. Furthermore, colocalization of 8OHG and OGG-1 with mitochondrial markers TOM 20 and aconitase, respectively, indicated significant levels of oxidatively-induced mtDNA damage and implicated a role for mitochondrial OGG-1 function. In vitro exposure of recombinant OGG-1 (rOGG-1) with increasing concentrations of 4HNE resulted in a concentration-dependent decrease in OGG-1 activity. Mass spectral analysis of trypsin digests of 4HNE-treated rOGG-1 identified 4HNE adducts on C28, C75, C163, H179, H237, C241, K249, H270, and H282. In silico molecular modeling of 4HNE-K249 OGG-1 and 4HNE-H270 OGG-1 mechanistically supported 4HNE-mediated enzymatic inhibition of OGG-1. In conclusion, these data support the hypothesis that inhibition of OGG-1 by direct modification by 4HNE contributes to decreased OGG-1 activity and increased 8OHG-modified DNA that are present in the diabetic heart.

4.
Adv Exp Med Biol ; 1193: 155-174, 2019.
Article in English | MEDLINE | ID: mdl-31368103

ABSTRACT

A major pathophysiological mechanism behind the development of diabetic heart diseases is oxidative stress mediated by toxic reactive aldehydes such as 4-hydroxynonenal (4HNE). Aldehyde dehydrogenase (ALDH) 2 is a mitochondrial enzyme that has been found to detoxify these deleterious aldehydes and thereby mitigate cardiac damage. Furthermore, its protective role in cellular signaling reverses aberrations caused by hyperglycemia, thereby protecting cardiac function. This chapter assesses the role of ALDH2 in diabetic heart diseases by examining preclinical studies where ALDH2 activity is perturbed in both decreased and increased directions. In doing so, issues in improving ALDH2 activity in select human populations are elucidated, and further research directions are discussed.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/genetics , Diabetes Mellitus/genetics , Heart Diseases/genetics , Aldehydes/adverse effects , Heart Diseases/complications , Humans , Oxidative Stress
5.
PLoS One ; 13(7): e0200697, 2018.
Article in English | MEDLINE | ID: mdl-30024944

ABSTRACT

The purpose of this study was to determine whether blocking of G protein ßγ (Gßγ) signaling halts heart failure (HF) progression by macrophage phenotype manipulation. Cardiac Gßγ signaling plays a crucial role in HF pathogenesis. Previous data suggested that inhibiting Gßγ signaling reprograms T helper cell 1 (Th1) and Th2 cytokines, suggesting that Gßγ might be a useful drug target for treating HF. We investigated the efficacy of a small molecule Gßγ inhibitor, gallein, in a clinically relevant, experimental autoimmune myocarditis (EAM) model of HF as well as in human macrophage phenotypes in vitro. In the myocardium of HF patients, we observed that G protein coupled receptor kinase (GRK)2 levels were down-regulated compared with healthy controls. In rat EAM, treatment with gallein effectively improved survival and cardiac function, suppressed cardiac remodeling, and further attenuated myocardial protein expression of GRK2 as well as high mobility group box (HMGB)1 and its cascade signaling proteins. Furthermore, gallein effectively inhibited M1 polarization and promoted M2 polarization in vivo in the EAM heart and in vitro in human monocyte-derived macrophages. Taken together, these data suggest that the small molecule Gßγ inhibitor, gallein, could be an important pharmacologic therapy for HF as it can switch the phenotypic reprogramming from M1 to M2 phenotype in a rat model of EAM heart and in human macrophages.


Subject(s)
Autoimmune Diseases/prevention & control , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Macrophages/drug effects , Myocarditis/prevention & control , Signal Transduction/drug effects , Xanthenes/pharmacology , Animals , Autoimmune Diseases/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , HMGB1 Protein/metabolism , Heart Failure/metabolism , Heart Failure/prevention & control , Humans , Macrophage Activation/drug effects , Macrophages/classification , Macrophages/metabolism , Male , Myocarditis/metabolism , Rats, Inbred Lew
6.
Nutrition ; 34: 47-54, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28063511

ABSTRACT

Diabetic cardiomyopathy (DCM) is described as impaired cardiac diastolic and systolic functions. Diabetes mellitus (DM), a related cardiovascular disease, has become one of the major causes of death in DM patients. Mortality in these diseases is 2 to 3 times higher than in non-DM patients with cardiovascular disease. The progression of DCM and the cellular and molecular perturbations associated with the pathogenesis are complex and multifactorial. Although considerable progress has been achieved, the molecular etiologies of DCM remain poorly understood. There is an expanding need for natural antidiabetic medicines that do not cause the side effects of modern drugs. Curcumin, a pleiotropic molecule, from Curcuma longa, is known to possess numerous impacts such as scavenging free radical, antioxidant, antitumor, and antiinflammatory activities. The reports from preclinical and clinical findings revealed that curcumin can reverse insulin resistance, hyperglycemia, obesity, and obesity-related metabolic diseases. The current review provides an updated overview of the possible molecular mechanism of DCM and multitarget approach of curcumin in alleviating DCM and diabetic complication. Additionally, we mentioned the approaches that are currently being implemented to improve the bioavailability of this promising natural product in diabetes therapeutics.


Subject(s)
Curcumin/pharmacokinetics , Diabetic Cardiomyopathies/drug therapy , Hypoglycemic Agents/pharmacokinetics , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Curcuma/chemistry , Curcumin/administration & dosage , Curcumin/chemistry , Disease Models, Animal , Heart/drug effects , Heart/physiology , Humans , Hypoglycemic Agents/administration & dosage , Oxidative Stress/drug effects , Plant Extracts/administration & dosage , Plant Extracts/pharmacokinetics
7.
Ageing Res Rev ; 35: 291-296, 2017 May.
Article in English | MEDLINE | ID: mdl-27825897

ABSTRACT

Because cardiovascular disease remains the major cause of mortality and morbidity world-wide, there remains a compelling need for new insights and novel therapeutic avenues. In this regard, the senescence-accelerated mouse prone 8 (SAMP8) line is a particularly good model for studying the effects of aging on cardiovascular health. Accumulating evidence suggests that this model may shed light on age-associated cardiac and vascular dysfunction and disease. These animals manifest evidence of inflammation, oxidative stress and adverse cardiac remodeling that may recapitulate processes involved in human disease. Early alterations in oxidative damage promote endoplasmic reticulum stress to trigger apoptosis and cytokine production in this genetically susceptible mouse strain. Conversely, pharmacological treatments that reduce inflammation and oxidative stress improve cardiac function in these animals. Therefore, the SAMP8 mouse model provides an exciting opportunity to expand our knowledge of aging in cardiovascular disease and the potential identification of novel targets of treatment. Herein, we review the previous studies performed in SAMP8 mice that provide insight into age-related cardiovascular alterations.


Subject(s)
Aging, Premature , Apoptosis/physiology , Cardiovascular System , Cellular Senescence/physiology , Inflammation/physiopathology , Oxidative Stress/physiology , Aging, Premature/metabolism , Aging, Premature/physiopathology , Animals , Cardiovascular System/metabolism , Cardiovascular System/physiopathology , Disease Models, Animal , Mice
8.
PLoS One ; 11(10): e0163158, 2016.
Article in English | MEDLINE | ID: mdl-27736868

ABSTRACT

Aldehyde dehydrogenase (ALDH) 2 is a mitochondrial isozyme of the heart involved in the metabolism of toxic aldehydes produced from oxidative stress. We hypothesized that hyperglycemia-mediated decrease in ALDH2 activity may impair mitochondrial respiration and ultimately result in cardiac damage. A single dose (65 mg/kg; i.p.) streptozotocin injection to rats resulted in hyperglycemia with blood glucose levels of 443 ± 9 mg/dl versus 121 ± 7 mg/dl in control animals, p<0.0001, N = 7-11. After 6 months of diabetes mellitus (DM) induction, the rats were sacrificed after recording the functionality of their hearts. Increase in the cardiomyocyte cross sectional area (446 ± 32 µm2 Vs 221 ± 10 µm2; p<0.0001) indicated cardiac hypertrophy in DM rats. Both diastolic and systolic dysfunctions were observed with DM rats compared to controls. Most importantly, myocardial ALDH2 activity and levels were reduced, and immunostaining for 4HNE protein adducts was increased in DM hearts compared to controls. The mitochondrial oxygen consumption rate (OCR), an index of mitochondrial respiration, was decreased in mitochondria isolated from DM hearts compared to controls (p<0.0001). Furthermore, the rate of mitochondrial respiration and the increase in carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP)-induced maximal respiration were also decreased with chronic hyperglycemia. Chronic hyperglycemia reduced mitochondrial OXPHOS proteins. Reduced ALDH2 activity was correlated with mitochondrial dysfunction, pathological remodeling and cardiac dysfunction, respectively. Our results suggest that chronic hyperglycemia reduces ALDH2 activity, leading to mitochondrial respiratory dysfunction and consequently cardiac damage and dysfunction.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/metabolism , Cardiomegaly/etiology , Cardiomegaly/pathology , Hyperglycemia/complications , Hyperglycemia/pathology , Mitochondria/pathology , Myocardium/pathology , Animals , Cardiomegaly/metabolism , Chronic Disease , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Hyperglycemia/metabolism , Male , Mitochondria/metabolism , Myocardium/metabolism , Oxidative Stress , Oxygen Consumption , Rats , Rats, Sprague-Dawley
9.
PLoS One ; 11(4): e0152922, 2016.
Article in English | MEDLINE | ID: mdl-27070323

ABSTRACT

The aim of this study was to investigate the role of macrophage polarization in aging heart. Macrophage differentiation is pathogenically linked to many inflammatory and immune disorders. It is often preceded by myocardial inflammation, which is characterized by increased cardiac damage and pro-inflammatory cytokine levels. Therefore, we investigated the hypothesis that senescence accelerated-prone (SAMP8) mice cardiac tissue would develop macrophage polarization compared with senescence-resistant control (SAMR1) mice. Both SAMP8 and SAMR1 mice were sacrificed when they became six month old. We evaluated, histo-pathological changes and modifications in protein expression by Western blotting and immuno-histochemical staining for M1 and M2 macrophage markers, high mobility group protein (HMG)B1 and its cascade proteins, pro-inflammatory factors and inflammatory cytokines in cardiac tissue. We observed significant upregulation of HMGB1, toll-like receptor (TLR)2, TLR4, nuclear factor (NF)κB p65, tumor necrosis factor (TNF)α, cyclooxygenase (COX)2, interferon (IFN)γ, interleukin (IL)-1ß, IL-6 and M1 like macrophage specific marker cluster of differentiation (CD)68 expressions in SAMP8 heart. In contrast, M2 macrophage specific marker CD36, and IL-10 expressions were down-regulated in SAMP8 mice. The results from the study demonstrated that, HMGB1-TLR2/TLR4 signaling cascade and induction of phenotypic switching to M1 macrophage polarization in SAMP8 mice heart would be one of the possible reasons behind the cardiac dysfunction and thus it could become an important therapeutic target to improve the age related cardiac dysfunction.


Subject(s)
Aging/metabolism , HMGB1 Protein/metabolism , Heart/physiology , Macrophages/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Ventricular Remodeling/physiology , Animals , Cyclooxygenase 2/metabolism , Inflammation/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice , NF-kappa B/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/physiology
10.
Biofactors ; 42(4): 368-75, 2016 Jul 08.
Article in English | MEDLINE | ID: mdl-27087487

ABSTRACT

Heart failure is typically related to aging as there is a definite relationship between age-related changes in the heart and the pathogenesis of heart failure. We have previously reported the involvement of p38 mitogen-activated protein kinase protein in cardiac function using animal models of heart failure. To further understand its relationship with aging-induced heart failure, we have compared its expression in the hearts of senescence accelerated-prone (SAMP8) mice and their control (SAMR1) with normal aging behavior. We have identified its activation along with reduced expression of 14-3-3η protein in SAMP8 mice hearts than in SAMR1 mice. To reveal the downstream signaling, we have measured the endoplasmic reticulum stress marker proteins along with some inflammatory and apoptosis markers and identified a significant increase in SAMP8 mice hearts than that of SAMR1. In addition, we have performed comet assay and revealed a significant DNA damage in the cardiomyocytes of SAMP8 mice when compared with SAMR1 mice. All these results demonstrate the role of 14-3-3η protein and the downstream mitogen-activated protein kinase-mediated endoplasmic reticulum stress, and apoptosis and DNA damage in aging-induced cardiac malfunction in SAMP8 mice. Thus targeting this signaling might be effective in treating age-related cardiac dysfunction. © 2016 BioFactors, 42(4):368-375, 2016.


Subject(s)
Aging , Endoplasmic Reticulum Stress , Myocardium/metabolism , 14-3-3 Proteins/metabolism , Animals , Apoptosis , Fibrosis , Heart Failure/metabolism , Heart Failure/pathology , MAP Kinase Signaling System , Male , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Myocardium/pathology , Myocytes, Cardiac/physiology , Oxidative Stress
11.
Alcohol Clin Exp Res ; 40(4): 686-97, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27013182

ABSTRACT

BACKGROUND: Eight percent of the U.S. population has been diagnosed with diabetes mellitus (DM), while another large percentage has gone undiagnosed. As the epidemiology of this disease constitutes a larger percentage of the American population, another factor presents a dangerous dilemma that can exacerbate the hazardous effects imposed by DM. Excessive alcohol consumption concerns the health of more than 50% of all adults. When this heavy-alcohol-drinking population overlaps with DM and its complications, the effects can be dangerous. In this review, we term it as "double trouble." METHODS: We provide evidence of alcohol-induced exacerbation of organ damage in diabetic conditions. In certain cases, we have explained how diabetes and alcohol induce similar pathological effects. RESULTS: Known exacerbated complications include those related to heart diseases, liver damage, kidney dysfunction, as well as retinal and neurological impairment. Often, pathophysiological damage concludes with end-stage disorders and even mortality. The metabolic, cell signaling, and pathophysiological changes associated with "double trouble" would lead to the identification of novel therapeutic targets. CONCLUSIONS: This review summarizes the epidemiology, diagnosis, pathophysiology, metabolic, and cell signaling alterations and finally brushes upon issues and strategies to manage the "double trouble."


Subject(s)
Alcohol Drinking/adverse effects , Alcohol Drinking/epidemiology , Diabetes Complications/epidemiology , Diabetes Mellitus/epidemiology , Alcohol Drinking/physiopathology , Alcoholism/diagnosis , Alcoholism/epidemiology , Alcoholism/physiopathology , Diabetes Complications/diagnosis , Diabetes Complications/physiopathology , Diabetes Mellitus/diagnosis , Diabetes Mellitus/physiopathology , Humans
12.
Cell Signal ; 28(2): 1-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26577527

ABSTRACT

Reactive oxygen species (ROS)-mediated reactive aldehydes induce cellular stress. In cardiovascular diseases such as ischemia-reperfusion injury, lipid-peroxidation derived reactive aldehydes such as 4-hydroxy-2-nonenal (4HNE) are known to contribute to the pathogenesis. 4HNE is involved in ROS formation, abnormal calcium handling and more importantly defective mitochondrial respiration. Aldehyde dehydrogenase (ALDH) superfamily contains NAD(P)(+)-dependent isozymes which can detoxify endogenous and exogenous aldehydes into non-toxic carboxylic acids. Therefore we hypothesize that 4HNE afflicts mitochondrial respiration and leads to cell death by impairing ALDH2 activity in cultured H9C2 cardiomyocyte cell lines. H9C2 cardiomyocytes were treated with 25, 50 and 75 µM 4HNE and its vehicle, ethanol as well as 25, 50 and 75 µM disulfiram (DSF), an inhibitor of ALDH2 and its vehicle (DMSO) for 4 h. 4HNE significantly decreased ALDH2 activity, ALDH2 protein levels, mitochondrial respiration and mitochondrial respiratory reserve capacity, and increased 4HNE adduct formation and cell death in cultured H9C2 cardiomyocytes. ALDH2 inhibition by DSF and ALDH2 siRNA attenuated ALDH2 activity besides reducing ALDH2 levels, mitochondrial respiration and mitochondrial respiratory reserve capacity and increased cell death. Our results indicate that ALDH2 impairment can lead to poor mitochondrial respiration and increased cell death in cultured H9C2 cardiomyocytes.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/metabolism , Mitochondria/enzymology , Myocytes, Cardiac/enzymology , Aldehyde Dehydrogenase, Mitochondrial/genetics , Aldehydes/toxicity , Animals , Cell Death , Cell Line , Cell Respiration/drug effects , DNA Adducts/metabolism , Disulfiram/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats
13.
Exp Biol Med (Maywood) ; 239(5): 610-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24651616

ABSTRACT

Reactive aldehydes such as 4-hydroxy-2-nonenal (4HNE) are generated in the myocardium in cardiac disease. 4HNE and other toxic aldehydes form adducts with proteins, leading to cell damage and organ dysfunction. Aldehyde dehydrogenases (ALDHs) metabolize toxic aldehydes such as 4HNE into nontoxic metabolites. Both ALDH levels and activity are reduced in cardiac disease. We examined whether reduced ALDH2 activity contributes to cardiomyocyte hypertrophy in mice fed a high-fat diet and injected with low-dose streptozotocin (STZ). These mice exhibited most of the characteristics of metabolic syndrome/type-2 diabetes mellitus (DM): increased blood glucose levels depicting hyperglycemia (415.2 ± 18.7 mg/dL vs. 265.2 ± 7.6 mg/dL; P < 0.05), glucose intolerance with normal plasma insulin levels, suggesting insulin resistance and obesity as evident from increased weight (44 ± 3.1 vs. 34.50 ± 1.32 g; P < 0.05) and body fat. Myocardial ALDH2 activity was 60% lower in these mice (0.1 ± 0.012 vs. 0.04 ± 0.015 µmol/min/mg protein; P < 0.05). Myocardial 4HNE levels were also elevated in the hyperglycemic hearts. Co-immunoprecipitation study showed that 4HNE formed adducts on myocardial ALDH2 protein in the mice exhibiting metabolic syndrome/type-2 DM, and they had obvious cardiac hypertrophy compared with controls as evident from increased heart weight (HW), HW to tibial length ratio, left ventricular (LV) mass and cardiomyocyte hypertrophy. Cardiomyocyte hypertrophy was correlated inversely with ALDH2 activity (R (2 )= 0.7; P < 0.05). Finally, cardiac dysfunction was observed in mice with metabolic syndrome/type-2 DM. Therefore, we conclude that reduced ALDH2 activity may contribute to cardiac hypertrophy and dysfunction in mice presenting with some of the characteristics of metabolic syndrome/type-2 DM when on a high-fat diet and low-dose STZ injection.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehydes/metabolism , Cardiomegaly/pathology , Cell Proliferation , Diet, High-Fat , Myocytes, Cardiac/physiology , Streptozocin/administration & dosage , Aldehyde Dehydrogenase, Mitochondrial , Animals , Diabetes Mellitus, Experimental/pathology , Male , Mice , Mice, Inbred C57BL
14.
J Cell Mol Med ; 16(9): 2176-85, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22268705

ABSTRACT

Edaravone, a novel antioxidant, acts by trapping hydroxyl radicals, quenching active oxygen and so on. Its cardioprotective activity against experimental autoimmune myocarditis (EAM) was reported. Nevertheless, it remains to be determined whether edaravone protects against cardiac remodelling in dilated cardiomyopathy (DCM). The present study was undertaken to assess whether edaravone attenuates myocardial fibrosis, and examine the effect of edaravone on cardiac function in rats with DCM after EAM. Rat model of EAM was prepared by injection with porcine cardiac myosin 28 days after immunization, we administered edaravone intraperitoneally at 3 and 10 mg/kg/day to rats for 28 days. The results were compared with vehicle-treated rats with DCM. Cardiac function, by haemodynamic and echocardiographic study and histopathology were performed. Left ventricular (LV) expression of NADPH oxidase subunits (p47(phox), p67(phox), gp91(phox) and Nox4), fibrosis markers (TGF-ß(1) and OPN), endoplasmic reticulum (ER) stress markers (GRP78 and GADD 153) and apoptosis markers (cytochrome C and caspase-3) were measured by Western blotting. Edaravone-treated DCM rats showed better cardiac function compared with those of the vehicle-treated rats. In addition, LV expressions of NADPH oxidase subunits levels were significantly down-regulated in edaravone-treated rats. Furthermore, the number of collagen-III positive cells in the myocardium of edaravone-treated rats was lower compared with those of the vehicle-treated rats. Our results suggest that edaravone ameliorated the progression of DCM by modulating oxidative and ER stress-mediated myocardial apoptosis and fibrosis.


Subject(s)
Antioxidants/pharmacology , Antipyrine/analogs & derivatives , Cardiomyopathy, Dilated/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antipyrine/pharmacology , Apoptosis/drug effects , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Blotting, Western , Cardiac Myosins , Cardiomyopathy, Dilated/pathology , Caspase 3/genetics , Caspase 3/metabolism , Cytochromes c/genetics , Cytochromes c/metabolism , Down-Regulation , Edaravone , Endomyocardial Fibrosis/drug therapy , Endomyocardial Fibrosis/pathology , Endoplasmic Reticulum Stress/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , In Situ Nick-End Labeling , Male , Myocarditis/drug therapy , Myocarditis/pathology , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Rats , Rats, Inbred Lew , Swine
15.
Free Radic Res ; 46(2): 154-63, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22145946

ABSTRACT

In order to test the hypothesis that treatment with quercetin at a dose of 10 mg/kg protects from the progression of experimental autoimmune myocarditis (EAM) to dilated cardiomyopathy (DCM), we have used the rat model of EAM induced by porcine cardiac myosin. Our results identified that the post-myocarditis rats suffered from elevated endoplasmic reticulum (ER) stress and adverse cardiac remodelling in the form of myocardial fibrosis, whereas the rats treated with quercetin have been protected from these changes as evidenced by the decreased myocardial levels of ER stress and fibrosis markers when compared with the vehicle-treated DCM rats. In addition, the myocardial dimensions and cardiac function were preserved significantly in the quercetin-treated rats in comparison with the DCM rats treated with vehicle alone. Interestingly, the rats treated with quercetin showed significant suppression of the myocardial endothelin-1 and also the mitogen activated protein kinases (MAPK) suggesting that the protection offered by quercetin treatment against progression of EAM involves the modulation of MAPK signalling cascade. Collectively, the present study provides data to support the role of quercetin in protecting the hearts of the rats with post myocarditis DCM.


Subject(s)
Autoimmune Diseases/drug therapy , Cardiotonic Agents/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endothelin-1/metabolism , MAP Kinase Signaling System , Myocarditis/drug therapy , Oxidative Stress/drug effects , Quercetin/pharmacology , Animals , Apoptosis/drug effects , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Biomarkers/metabolism , Cardiac Myosins , Cardiotonic Agents/therapeutic use , Cytochromes c/metabolism , Fibrosis , Heart/drug effects , Male , Myocarditis/chemically induced , Myocarditis/immunology , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology , Organ Size/drug effects , Osteopontin/metabolism , Quercetin/therapeutic use , Rats , Rats, Inbred Lew , Ventricular Remodeling/drug effects
16.
Toxicology ; 291(1-3): 139-45, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22120037

ABSTRACT

Candesartan cilexetil, an angiotensin (Ang) II receptor 1 blocker was reported to suppress the myocardial damage in various cardiovascular complications but the mode by which it is effective in preventing the progression of dilated cardiomyopathy (DCM) is unknown. Emerging evidences suggest that, at least, part of the benefits observed with the use of AT1 receptor blockers could be attributed to the increased Ang (1-7) levels observed during administration of these agents. Identification of the novel components of the RAS, ACE2 and Ang (1-7) receptor mas, provided essential elements for considering the existence of a vasodilator arm of the RAS, represented by the ACE2-Ang (1-7)-mas axis. In this study, rat model of DCM was prepared by injection with porcine cardiac myosin. Twenty-eight days after immunization, candesartan cilexetil was administered intraperitoneally at 1 or 10mg/kg/day to rats for four weeks. Myocardial expression of Ang receptors and markers of calcium homeostasis, endoplasmic reticulum (ER) stress and apoptosis were measured by Western blotting and histopathological staining techniques. Candesartan improved the functional markers in a dose-dependent manner and also upregulated Ang (1-7), ACE2 and mas1 in the myocardium of DCM rats. Various ER stress and apoptosis markers were attenuated and the number apoptotic cells were significantly lower in the candesartan treated rats compared with those of the vehicle group. These findings suggest that candesartan treatment prevented the progression of DCM by activation of the counter regulatory arm of the RAS and possibly through modulation of ER stress and subsequently, cardiac apoptosis.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin I/pharmacology , Apoptosis/drug effects , Benzimidazoles/pharmacology , Biphenyl Compounds/pharmacology , Endoplasmic Reticulum Stress/drug effects , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Myosins/antagonists & inhibitors , Myosins/toxicity , Peptide Fragments/pharmacology , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Proteins/physiology , Receptors, G-Protein-Coupled/physiology , Tetrazoles/pharmacology , Angiotensin II/biosynthesis , Angiotensin-Converting Enzyme 2 , Animals , Apoptosis Regulatory Proteins/biosynthesis , Autoimmune Diseases/chemically induced , Autoimmune Diseases/prevention & control , Blotting, Western , Cell Survival/drug effects , Immunohistochemistry , In Situ Nick-End Labeling , Male , Myocarditis/chemically induced , Myocarditis/prevention & control , Proto-Oncogene Mas , Rats , Rats, Inbred Lew , Sarcoplasmic Reticulum Calcium-Transporting ATPases/biosynthesis , Signal Transduction/physiology
17.
Cell Physiol Biochem ; 28(5): 911-22, 2011.
Article in English | MEDLINE | ID: mdl-22178943

ABSTRACT

Diabetic cardiomyopathy is associated with increased oxidative stress and inflammation. Mammalian 14-3-3 proteins are dimeric phosphoserine-binding proteins that participate in signal transduction and regulate several aspects of cellular biochemistry. The aim of the study presented here was to clarify the role of 14-3-3 protein in the mitogen activated protein kinase (MAPK) and nuclear factor-kB (NF-κB) signaling pathway after experimental diabetes by using transgenic mice with cardiac-specific expression of a dominant-negative 14-3-3 protein mutant (DN 14-3-3). Significant p-p38 MAPK activation in DN 14-3-3 mice compared to wild type mice (WT) after diabetes induction and with a corresponding up regulation of its downstream effectors, p-MAPK activated protein kinase 2 (MAPKAPK-2). Marked increases in cardiac hypertrophy, fibrosis and inflammation were observed with a corresponding up-regulation of atrial natriuretic peptide, osteopontin, connective tissue growth factor, tumor necrosis factor α, interleukin (IL)-1ß, IL-6 and cellular adhesion molecules. Moreover, reactive oxygen species, left ventricular expression of NADPH oxidase subunits, p22 phox, p67 phox, and Nox4, and lipid peroxidation levels were significantly increased in diabetic DN 14-3-3mice compared to diabetic WT mice. Furthermore, myocardial NF-κB activation, inhibitor of kappa B-α degradation and mRNA expression of proinflammatory cytokines were significantly increased in DN 14-3-3 mice compared to WT mice after diabetes induction. In conclusion, our data suggests that depletion of 14-3-3 protein induces cardiac oxidative stress, inflammation and remodeling after experimental diabetes induction mediated through p38 MAPK, MAPKAPK-2 and NF-κB signaling.


Subject(s)
Diabetes Mellitus, Experimental , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Oxidative Stress , Signal Transduction , Ventricular Remodeling , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Animals , Cardiomegaly/metabolism , Cell Adhesion Molecules/metabolism , Cytokines/genetics , Cytokines/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Gene Expression Regulation , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Transgenic , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Streptozocin , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Eur J Pharm Sci ; 44(5): 627-34, 2011 Dec 18.
Article in English | MEDLINE | ID: mdl-22033153

ABSTRACT

There is evidence that the activation of renal angiotensin (Ang)-II plays a critical role in the pathogenesis of diabetic kidney diseases (DN) via the ER stress-induced renal apoptosis. Since, the potential negative role of Ang-II in the pathogenesis of ER stress-mediated apoptosis is poorly understood; we evaluated whether treatment of mice with AT-1R specific blocker, olmesartan is associated with the reduction of ER stress-induced renal apoptosis in streptozotocin (STZ)-induced diabetic animal model. We employed western blot analysis to measure the renal protein expressions level of NADPH oxidase subunits, ER chaperone GRP78 and the ER-associated apoptosis proteins. Furthermore, TUNEL staining was used to measure the renal apoptosis. Additionally, dihydroethidium staining and TBARS assay, and immunohistochemistry were performed to measure the renal superoxide radical production and lipid peroxidation, and activation of an Ang-II, respectively. The diabetic kidney mice were found to have increased protein expressions of NADPH oxidase subunits, GRP78 and ER-associated apoptosis proteins, such as TRAF2, IRE-1α, CHOP, p-JNK and procaspase-12, in comparison to normal mice, and which were significantly blunted by the olmesartan treatment in diabetic kidney mice. Furthermore, the diabetic kidney mice were found to have significant increment in renal apoptosis, superoxide radical production, MDA level and activation of an Ang-II and which were also attenuated by the olmesartan treatment. Considering all the findings, it is suggested that the AT-1R specific blocker-olmesartan treatment could be a potential therapy in treating ER stress-induced renal apoptosis via the modulation of AT-1R/CHOP-JNK-Caspase12 pathway in STZ-induced diabetic mice.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Apoptosis/drug effects , Diabetes Mellitus, Experimental/drug therapy , Endoplasmic Reticulum/drug effects , Imidazoles/therapeutic use , Kidney/drug effects , Tetrazoles/therapeutic use , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Blood Glucose/analysis , Caspase 12/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney/metabolism , Lipid Peroxidation/drug effects , Malondialdehyde/metabolism , Membrane Proteins/metabolism , Mice , Olmesartan Medoxomil , Protein Serine-Threonine Kinases/metabolism , Receptor, Angiotensin, Type 1/metabolism , Superoxides/metabolism , TNF Receptor-Associated Factor 2/metabolism , Tetrazoles/pharmacology , Transcription Factor CHOP/metabolism
19.
J Mol Cell Cardiol ; 51(6): 980-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21920368

ABSTRACT

We previously found that in the hearts of hypertensive Dahl salt-sensitive rats, ßIIPKC levels increase during the transition from compensated cardiac hypertrophy to cardiac dysfunction. Here we showed that a six-week treatment of these hypertensive rats with a ßIIPKC-specific inhibitor, ßIIV5-3, prolonged their survival by at least 6weeks, suppressed myocardial fibrosis and inflammation, and delayed the transition from compensated hypertrophy to cardiac dysfunction. In addition, changes in the levels of the Ca(2+)-handling proteins, SERCA2 and the Na(+)/Ca(2+) exchanger, as well as troponin I phosphorylation, seen in the control-treated hypertensive rats were not observed in the ßΙΙPKC-treated rats, suggesting that ßΙΙPKC contributes to the regulation of calcium levels in the myocardium. In contrast, treatment with the selective inhibitor of ßIPKC, an alternative spliced form of ßIIPKC, had no beneficial effects in these rats. We also found that ßIIV5-3, but not ßIV5-3, improved calcium handling in isolated rat cardiomyocytes and enhanced contractility in isolated rat hearts. In conclusion, our data using an in vivo model of cardiac dysfunction (late-phase hypertrophy), suggest that ßIIPKC contributes to the pathology associated with heart failure and thus an inhibitor of ßIIPKC may be a potential treatment for this disease.


Subject(s)
Cardiomegaly/enzymology , Cardiomegaly/prevention & control , Peptides/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Amino Acid Sequence , Animals , Calcium/metabolism , Cardiomegaly/mortality , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Disease Models, Animal , Fibrosis , Heart Failure/metabolism , Heart Failure/mortality , Heart Failure/prevention & control , Male , Myocardial Contraction/drug effects , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Protein Kinase C beta , Rats , Rats, Inbred Dahl , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Calcium Exchanger/metabolism
20.
Drug Discov Today ; 15(19-20): 826-41, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20708094

ABSTRACT

Arginine vasopressin (AVP) attracted attention as a potentially important neurohormonal mediator of the heart failure (HF) syndrome and hyponatremic states in humans because AVP influences renal handling of free water, vasoconstriction and myocyte biology through activation of V2 and V1(a) receptors. Current research is exploring V2- and dual V1(a)/V2 receptor antagonism for the treatment of hyponatremia, as well as for the congestion and edema associated with chronic HF, because vasopressin receptor antagonists might offer benefits in comparison with conventional loop diuretics. The purpose of this review is to update the current status of experimental and clinical studies with available vasopressin receptor antagonists (conivaptan and tolvaptan) and their potential role in the treatment of HF and hyponatremia of multiple causes.


Subject(s)
Antidiuretic Hormone Receptor Antagonists , Benzazepines/pharmacology , Heart Failure/drug therapy , Hyponatremia/drug therapy , Arginine Vasopressin/metabolism , Benzazepines/therapeutic use , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Hyponatremia/etiology , Receptors, Vasopressin/metabolism , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Tolvaptan
SELECTION OF CITATIONS
SEARCH DETAIL
...