Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Can J Physiol Pharmacol ; 102(1): 14-25, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37748207

ABSTRACT

Cardiovascular disease (CVD) complications have remained a major cause of death among patients with diabetes. Hence, there is a need for effective therapeutics against diabetes-induced CVD complications. Since its discovery, proprotein convertase subtilisin/kexin type 9 (PCSK9) has been reported to be involved in the pathology of various CVDs, with studies showing a positive association between plasma levels of PCSK9, hyperglycemia, and dyslipidemia. PCSK9 regulates lipid homeostasis by interacting with low-density lipoprotein receptors (LDLRs) present in hepatocytes and subsequently induces LDLR degradation via receptor-mediated endocytosis, thereby reducing LDL uptake from circulation. In addition, PCSK9 also induces pro-inflammatory cytokine expression and apoptotic cell death in diabetic-CVD. Furthermore, therapies designed to inhibit PCSK9 effectively reduces diabetic dyslipidemia with clinical studies reporting reduced cardiovascular events in patients with diabetes and no significant adverse effect on glycemic controls. In this review, we discuss the role of PCSK9 in the pathogenesis of diabetes-induced CVD and the potential mechanisms by which PCSK9 inhibition reduces cardiovascular events in diabetic patients.


Subject(s)
Cardiovascular Diseases , Diabetes Complications , Diabetes Mellitus , Dyslipidemias , Humans , Proprotein Convertase 9/metabolism , Proprotein Convertase 9/therapeutic use , Dyslipidemias/complications , Dyslipidemias/drug therapy , Dyslipidemias/metabolism , Cardiovascular Diseases/etiology , Subtilisins/therapeutic use , Diabetes Mellitus/drug therapy
2.
Heart Fail Rev ; 26(3): 699-709, 2021 05.
Article in English | MEDLINE | ID: mdl-33033908

ABSTRACT

Vitamin C (Vit C) is an ideal antioxidant as it is easily available, water soluble, very potent, least toxic, regenerates other antioxidants particularly Vit E, and acts as a cofactor for different enzymes. It has received much attention due to its ability in limiting reactive oxygen species, oxidative stress, and nitrosative stress, as well as it helps to maintain some of the normal metabolic functions of the cell. However, over 140 clinical trials using Vit C in different pathological conditions such as myocardial infarction, gastritis, diabetes, hypertension, stroke, and cancer have yielded inconsistent results. Such a divergence calls for new strategies to establish practical significance of Vit C in heart failure or even in its prevention. For a better understanding of Vit C functioning, it is important to revisit its transport across the cell membrane and subcellular interactions. In this review, we have highlighted some historical details of Vit C and its transporters in the heart with a particular focus on heart failure in cancer chemotherapy.


Subject(s)
Ascorbic Acid , Heart Failure , Antioxidants/therapeutic use , Heart Failure/drug therapy , Humans , Oxidative Stress , Reactive Oxygen Species
3.
J Nutr ; 150(9): 2353-2363, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32510147

ABSTRACT

BACKGROUND: Although the combination of doxorubicin (DOX) and trastuzumab (TRZ) reduces the progression and recurrence of breast cancer, these anticancer drugs are associated with significant cardiotoxic side effects. OBJECTIVE: We investigated whether prophylactic administration of flaxseed (FLX) and its bioactive components, α-linolenic acid (ALA) and secoisolariciresinol diglucoside (SDG), would be cardioprotective against DOX + TRZ-mediated cardiotoxicity in a chronic in vivo female murine model. METHODS: Wild-type C57BL/6 female mice (10-12 wk old) received daily prophylactic treatment with one of the following diets: 1) regular control (RC) semi-purified diet; 2) 10% FLX diet; 3) 4.4% ALA diet; or 4) 0.44% SDG diet for a total of 6 wks. Within each arm, mice received 3 weekly injections of 0.9% saline or a combination of DOX [8 mg/(kg.wk)] and TRZ [3 mg/(kg.wk)] starting at the end of week 3. The main outcome was to evaluate the effects of FLX, ALA, and SDG on cardiovascular remodeling and markers of apoptosis, inflammation, and mitochondrial dysfunction. Significance between measurements was determined using a 4 (diet) × 2 (chemotherapy) × 2 (time) mixed factorial design with repeated measures. RESULTS: In the RC + DOX + TRZ-treated mice at week 6 of the study, the left ventricular ejection fraction (LVEF) decreased by 50% compared with the baseline LVEF (P < 0.05). However, the prophylactic administration of the FLX, ALA, or SDG diet was partially cardioprotective, with mice in these treatment groups showing an ∼68% increase in LVEF compared with the RC + DOX + TRZ-treated group at week 6 (P < 0.05). Although markers of inflammation (nuclear transcription factor κB), apoptosis [poly (ADP-ribose) polymerase-1 and the ratio of BCL2-associated X protein to B-cell lymphoma-extra large], and mitochondrial dysfunction (BCL2-interacting protein 3) were significantly elevated by approximately 2-fold following treatment with RC + DOX + TRZ compared with treatment with RC + saline at week 6, prophylactic administration of FLX, ALA, or SDG partially downregulated these signaling pathways. CONCLUSION: In a chronic in vivo female C57BL/6 mouse model of DOX + TRZ-mediated cardiotoxicity, FLX, ALA, and SDG were partially cardioprotective.


Subject(s)
Dietary Supplements , Doxorubicin/adverse effects , Flax , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Trastuzumab/adverse effects , Animals , Antineoplastic Agents/adverse effects , Cardiotoxicity , Female , Mice , Mice, Inbred C57BL , Ventricular Function, Left
4.
Am J Physiol Heart Circ Physiol ; 316(3): H435-H445, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30525893

ABSTRACT

Among the different cardiovascular disease complications, atherosclerosis-induced myocardial infarction (MI) is the major contributor of heart failure (HF) and loss of life. This review presents short- and long-term features of post-MI in human hearts and animal models. It is known that the heart does not regenerate, and thus loss of cardiac cells after an MI event is permanent. In survivors of a heart attack, multiple neurohumoral adjustments as well as simultaneous remodeling in both infarcted and noninfarcted regions of the heart help sustain pump function post-MI. In the early phase, migration of inflammatory cells to the infarcted area helps repair and remove the cell debris, while apoptosis results in the elimination of damaged cardiomyocytes, and there is an increase in the antioxidant response to protect the survived myocardium against oxidative stress (OS) injury. However, in the late phase, it appears that there is a relative increase in OS and activation of the innate inflammatory response in cardiomyocytes without any obvious inflammatory cells. In this late stage in survivors of MI, a progressive slow activation of these processes leads to apoptosis, fibrosis, cardiac dysfunction, and HF. Thus, this second phase of an increase in OS, innate inflammatory response, and apoptosis results in wall thinning, dilatation, and consequently HF. It is important to note that this inflammatory response appears to be innate to cardiomyocytes. Blunting of this innate immune cardiomyocyte response may offer new hope for the management of HF.


Subject(s)
Heart Failure/immunology , Immunity, Innate , Myocardial Infarction/immunology , Animals , Apoptosis , Heart Failure/etiology , Humans , Myocardial Infarction/etiology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Oxidative Stress
5.
Am J Physiol Heart Circ Physiol ; 316(3): H446-H458, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30499710

ABSTRACT

Although anticancer systemic therapy agents clearly lead to improved survival in patients with cancer, these can come at the cost of serious complications including cardiotoxicity. Two types of targeted systemic therapies currently in use for colorectal cancer (CRC) and renal cell cancer (RCC), respectively, include the vascular endothelial growth factor inhibitor bevacizumab (BVZ) and the tyrosine kinase inhibitor sunitinib (SNT). Despite the beneficial effects of BVZ and SNT in improving clinical outcomes in the settings of CRC and RCC, there is an increased risk of cardiac dysfunction. The aim of the present study was to determine whether prophylactic administration of renin-angiotensin system (RAS) inhibitors would attenuate the cardiotoxic side effects of BVZ or SNT in a chronic in vivo murine model. A total of 194 wild-type C57Bl/6 male mice received: 1) 0.9% saline, 2) BVZ (10 mg·kg-1·wk-1), or 3) SNT (40 mg·kg-1·day-1) for 4 wk. Within each arm, mice received daily prophylactic treatment with hydralazine (0.05 mg/ml), aliskiren (50 mg/kg), perindopril (4 mg/kg), or valsartan (2 mg/kg). Although hydralazine effectively lowered blood pressure in BVZ- or SNT-treated mice, it did not prevent left ventricular systolic dysfunction. Prophylactic administration of aliskiren, perindopril, or valsartan prevented adverse cardiovascular remodeling in mice treated with either BVZ or SNT. The addition of RAS antagonists also downregulated expression of phosphorylated p38 and Bcl-2-like 19-kDa interacting protein 3 in SNT-treated mice. In our chronic in vivo murine model, RAS antagonists partially attenuated the development of BVZ- or SNT-mediated cardiac dysfunction. Future clinical studies are warranted to investigate the cardioprotective effects of prophylactic treatment with RAS inhibitors in the settings of CRC and RCC. NEW & NOTEWORTHY In the evolving field of cardio-oncology, bevacizumab and sunitinib improve clinical outcomes in the settings of metastatic colorectal cancer and renal cell cancer, respectively. These anticancer drugs, however, are associated with an increased risk of cardiotoxicity. The prophylactic administration of renin-angiotensin system antagonists is partially cardioprotective against bevacizumab- and sunitinib-mediated cardiac dysfunction.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Antihypertensive Agents/therapeutic use , Antineoplastic Agents/toxicity , Renin-Angiotensin System , Ventricular Dysfunction/prevention & control , Amides/administration & dosage , Amides/therapeutic use , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Antihypertensive Agents/administration & dosage , Bevacizumab/toxicity , Cardiotoxicity , Fumarates/administration & dosage , Fumarates/therapeutic use , Hydralazine/administration & dosage , Hydralazine/therapeutic use , Male , Mice , Mice, Inbred C57BL , Perindopril/administration & dosage , Perindopril/therapeutic use , Sunitinib/toxicity , Valsartan/administration & dosage , Valsartan/therapeutic use , Ventricular Dysfunction/drug therapy , Ventricular Dysfunction/etiology
6.
Can J Physiol Pharmacol ; 97(9): 880-884, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31365282

ABSTRACT

The objective of this study was to analyze the cardioprotective roles of 3 wild blueberry genotypes and one commercial blueberry genotype by measuring markers of oxidative stress and cell death in H9c2 cardiac cells exposed to doxorubicin. Ripe berries of the 3 wild blueberry genotypes were collected from a 10-year-old clearcut forest near Nipigon, Ontario, Canada (49°1'39″N, 87°52'21″W), whereas the commercial blueberries were purchased from a local grocery store. H9c2 cardiac cells were incubated with 15 µg gallic acid equivalent/mL blueberry extract for 4 h followed by 5 µM doxorubicin for 4 h, and oxidative stress and active caspase 3/7 were analyzed. The surface area as well as total phenolic content was significantly higher in all 3 wild blueberry genotypes compared with the commercial species. Increase in oxidative stress due to doxorubicin exposure was attenuated by pre-treatment with all 3 types of wild blueberries but not by commercial berries. Furthermore, increase in caspase 3/7 activity was also attenuated by all 3 wild genotypes as well. These data demonstrate that wild blueberry extracts can attenuate doxorubicin-induced damage to H9c2 cardiomyocytes through reduction in oxidative stress and apoptosis, whereas the commercial blueberry had little effect.


Subject(s)
Blueberry Plants/chemistry , Cytoprotection/drug effects , Doxorubicin/adverse effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Plant Extracts/pharmacology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Phenols/analysis , Plant Extracts/chemistry , Rats
7.
Can J Physiol Pharmacol ; 97(4): 287-292, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30543459

ABSTRACT

Uncontrolled production of oxygen and nitrogen radicals results in oxidative and nitrosative stresses that impair cellular functions and have been regarded as causative common denominators of many pathological processes. In this review, we report on the beneficial effects of molecular hydrogen in scavenging radicals in an artificial system of •OH formation. As a proof of principle, we also demonstrate that in rat hearts in vivo, administration of molecular hydrogen led to a significant increase in superoxide dismutase as well as pAKT, a cell survival signaling molecule. Irradiation of the rats caused a significant increase in lipid peroxidation, which was mitigated by pre-treatment of the animals with molecular hydrogen. The nuclear factor erythroid 2-related factor 2 is regarded as an important regulator of oxyradical homeostasis, as well as it supports the functional integrity of cells, particularly under conditions of oxidative stress. We suggest that the beneficial effects of molecular hydrogen may be through the activation of nuclear factor erythroid 2-related factor 2 pathway that promotes innate antioxidants and reduction of apoptosis, as well as inflammation.


Subject(s)
Free Radical Scavengers/pharmacology , Hydrogen/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Radiation Injuries/metabolism , Animals , Humans , Hydroxyl Radical/metabolism
8.
Am J Physiol Cell Physiol ; 312(4): C418-C427, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28100487

ABSTRACT

An increase in oxidative stress is suggested to be the main cause in Doxorubicin (Dox)-induced cardiotoxicity. However, there is now evidence that activation of inducible nitric oxide synthase (iNOS) and nitrosative stress are also involved. The role of vitamin C (Vit C) in the regulation of nitric oxide synthase (NOS) and reduction of nitrosative stress in Dox-induced cardiotoxicity is unknown. The present study investigated the effects of Vit C in the mitigation of Dox-induced changes in the levels of nitric oxide (NO), NOS activity, protein expression of NOS isoforms, and nitrosative stress as well as cytokines TNF-α and IL-10 in isolated cardiomyocytes. Cardiomyocytes isolated from adult Sprague-Dawley rats were segregated into four groups: 1) control, 2) Vit C (25 µM), 3) Dox (10 µM), and 4) Vit C + Dox. Dox caused a significant increase in the generation of superoxide radical (O2·-), peroxynitrite, and NO, and these effects of Dox were blunted by Vit C. Dox increased the expression of iNOS and altered protein expression as well as activation of endothelial NOS (eNOS). These changes were prevented by Vit C. Dox induced an increase in the ratio of monomeric/dimeric eNOS, promoting the production of O2·-, which was prevented by Vit C by increasing the stability of the dimeric form of eNOS. Vit C protected against the Dox-induced increase in TNFα as well as a reduction in IL-10. These results suggest that Vit C provides cardioprotection by reducing oxidative/nitrosative stress and inflammation via a modulation of Dox-induced increase in the NO levels and NOS activity.


Subject(s)
Ascorbic Acid/metabolism , Doxorubicin/administration & dosage , Myocytes, Cardiac/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Oxidative Stress/physiology , Animals , Antibiotics, Antineoplastic/administration & dosage , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Male , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
9.
Am J Physiol Heart Circ Physiol ; 312(6): H1238-H1247, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28432060

ABSTRACT

It has been suggested that Toll-like receptor (TLR)4 promotes IL-10-mediated cardiac cell survival, whereas another receptor, TLR2, from the same family, is detrimental. Here, we examined the interactive role of these two innate signaling molecules under stressful conditions, including IL-10 knockout (IL-10-/-) mice, global ischemia-reperfusion (I/R) injury in rat hearts, and in vitro short hairpin RNA experimental models in the presence or absence of IL-10 (10 ng/ml). Circulating and myocardial levels of TNF-α as well as apoptosis and fibrosis were higher in IL-10-/- mice. The increase in TLR2 in IL-10-/- hearts indicated its negative regulation by IL-10. Ex vivo I/R also caused a marked upregulation of TLR2 and TNF-α as well as apoptotic and fibrotic signals. However, a 40-min reperfusion with IL-10 triggered an increase in TLR4 expression and improved recovery of cardiac function. The increase in IL-1 receptor-associated kinase (IRAK)-M and IRAK-2 activity during I/R injury suggested their role in TLR2 signaling. In vitro inhibition of TLR4 activity as a consequence of RNA inhibition-mediated suppression of myeloid differentiation gene (MyD)88 suggested MyD88-dependent activation of TLR4. The inclusion of IL-10 during reperfusion also downregulated the expression of IRAK-2, TNF-α receptor-associated factor 1-interacting protein (TRAIP) and apoptotic signals, caspase-3, and the Bax-to-Bcl-xL ratio. IL-10 reduced the TNF-α receptor-associated increase in TRAIP-induced apoptosis during I/R injury, which led to an increase in IL-1ß to mitigate transforming growth factor-ß receptor type I-mediated fibrosis. The IL-10 mitigation of these changes suggests that the stimulation through TLR4 signaling promotes IRAK-4 and phosphorylates IRAK-1 instead of IRAK-2 and may be an important therapeutic approach in restoring heart health in stress.NEW & NOTEWORTHY Under stress conditions such as downregulation of the IL-10 gene or ischemia-reperfusion injury, Toll-like receptor (TLR)4 and IL-1 receptor-associated kinase (IRAK)-1 activation is suppressed, along with the upregulation of TLR-2 and IRAK-2, resulting in fibrosis and apoptosis. It is suggested that IL-10 helps to maintain heart function during stress via myeloid differentiation gene 88/IRAK-4/IRAK-1-dependent TLR4 signaling.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Fibrosis , Genotype , Inflammation Mediators/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Phenotype , Phosphorylation , RNA Interference , Rats, Sprague-Dawley , Signal Transduction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Transfection , Ventricular Function, Left
10.
Am J Physiol Heart Circ Physiol ; 313(4): H795-H809, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28710069

ABSTRACT

Increase in oxidative/nitrosative stress is one of the mechanisms associated with the development of cardiotoxicity due to doxorubicin (Dox), a potent chemotherapy drug. Previously, we reported mitigation of Dox-induced oxidative/nitrosative stress and apoptosis by vitamin C (Vit C) in isolated cardiomyocytes. In the present in vivo study in rats, we investigated the effect of prophylactic treatment with Vit C on Dox-induced apoptosis, inflammation, oxidative/nitrosative stress, cardiac dysfunction, and Vit C transporter proteins. Dox (cumulative dose: 15 mg/kg) in rats reduced systolic and diastolic cardiac function and caused structural damage. These changes were associated with a myocardial increase in reactive oxygen species, reduction in antioxidant enzyme activities, increased expression of apoptotic proteins, and inflammation. Dox also caused an increase in the expression of proapoptotic proteins Bax, Bnip-3, Bak, and caspase-3. An increase in oxidative/nitrosative stress attributable to Dox was indicated by an increase in superoxide, protein carbonyl formation, lipid peroxidation, nitric oxide (NO), NO synthase (NOS) activity, protein nitrosylation, and inducible NOS protein expression. Dox increased the levels of cardiac proinflammatory cytokines TNF-α, IL-1ß, and IL-6, whereas the expression of Vit C transporter proteins (sodium-ascorbate cotransporter 2 and glucose transporter 4) was reduced. Prophylactic and concurrent treatment with Vit C prevented all these changes and improved survival in the Vit C + Dox group. Vit C also improved Dox-mediated systolic and diastolic dysfunctions and structural damage. These results suggest a cardioprotective role of Vit C in Dox-induced cardiomyopathy by reducing oxidative/nitrosative stress, inflammation, and apoptosis, as well as improving Vit C transporter proteins.NEW & NOTEWORTHY This in vivo study provides novel data that vitamin C improves cardiac structure and function in doxorubicin-induced cardiomyopathy by reducing oxidative/nitrosative stress, apoptosis, and inflammation along with upregulation of cardiac vitamin C transporter proteins. The latter may have a crucial role in improving antioxidant status in this cardiomyopathy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibiotics, Antineoplastic , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Cardiomyopathies/chemically induced , Cardiomyopathies/drug therapy , Cardiotonic Agents/pharmacology , Doxorubicin , Oxidative Stress/drug effects , Stress, Physiological/drug effects , Animals , Cytokines/biosynthesis , Electrocardiography/drug effects , Male , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/metabolism , Oxygen Consumption/drug effects , Rats , Rats, Wistar , Reactive Nitrogen Species , Survival Analysis
11.
Can J Physiol Pharmacol ; 95(10): 1078-1090, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28187269

ABSTRACT

A rapid rise in obesity, as well as physical inactivity, in industrialized countries is associated with fructose-consumption-mediated metabolic syndrome having a strong association with cardiovascular disease. Although insulin resistance is thought to be at the core, visceral obesity, hypertension, and hypertriglyceridemia are also considered important components of this metabolic disorder. In addition, various other abnormalities such as inflammation, oxidative stress, and elevated levels of uric acid are also part of this syndrome. Lifestyle changes through improved physical activity, as well as nutrition, are important approaches to minimize metabolic syndrome and its deleterious effects.


Subject(s)
Dietary Sugars/adverse effects , Fructose/adverse effects , Inflammation/etiology , Metabolic Syndrome/etiology , Oxidative Stress , Animals , Blood Pressure , Healthy Lifestyle , Humans , Hypertension/blood , Hypertension/etiology , Hypertension/physiopathology , Hyperuricemia/blood , Hyperuricemia/etiology , Hyperuricemia/metabolism , Inflammation/blood , Inflammation/physiopathology , Inflammation/therapy , Inflammation Mediators/blood , Metabolic Syndrome/blood , Metabolic Syndrome/physiopathology , Metabolic Syndrome/therapy , Prognosis , Risk Factors , Risk Reduction Behavior , Uric Acid/blood
12.
Can J Physiol Pharmacol ; 95(10): 1190-1203, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28750189

ABSTRACT

Irradiation of normal tissues leads to acute increase in reactive oxygen/nitrogen species that serve as intra- and inter-cellular signaling to alter cell and tissue function. In the case of chest irradiation, it can affect the heart, blood vessels, and lungs, with consequent tissue remodelation and adverse side effects and symptoms. This complex process is orchestrated by a large number of interacting molecular signals, including cytokines, chemokines, and growth factors. Inflammation, endothelial cell dysfunction, thrombogenesis, organ dysfunction, and ultimate failing of the heart occur as a pathological entity - "radiation-induced heart disease" (RIHD) that is major source of morbidity and mortality. The purpose of this review is to bring insights into the basic mechanisms of RIHD that may lead to the identification of targets for intervention in the radiotherapy side effect. Studies of authors also provide knowledge about how to select targeted drugs or biological molecules to modify the progression of radiation damage in the heart. New prospective studies are needed to validate that assessed factors and changes are useful as early markers of cardiac damage.


Subject(s)
Coronary Vessels/radiation effects , Heart Diseases/etiology , Inflammation Mediators/metabolism , Myocytes, Cardiac/radiation effects , Radiation Injuries/etiology , Reactive Oxygen Species/metabolism , Animals , Apoptosis/radiation effects , Biomarkers/metabolism , Coronary Vessels/metabolism , Coronary Vessels/pathology , DNA Damage , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/radiation effects , Heart Diseases/metabolism , Heart Diseases/pathology , Humans , Lipid Peroxidation/radiation effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/radiation effects , Radiation Injuries/metabolism , Radiation Injuries/pathology , Signal Transduction/radiation effects
13.
Heart Fail Rev ; 21(1): 11-23, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26542377

ABSTRACT

Sedentary life style and high calorie dietary habits are prominent leading cause of metabolic syndrome in modern world. Obesity plays a central role in occurrence of various diseases like hyperinsulinemia, hyperglycemia and hyperlipidemia, which lead to insulin resistance and metabolic derangements like cardiovascular diseases (CVDs) mediated by oxidative stress. The mortality rate due to CVDs is on the rise in developing countries. Insulin resistance (IR) leads to micro or macro angiopathy, peripheral arterial dysfunction, hampered blood flow, hypertension, as well as the cardiomyocyte and the endothelial cell dysfunctions, thus increasing risk factors for coronary artery blockage, stroke and heart failure suggesting that there is a strong association between IR and CVDs. The plausible linkages between these two pathophysiological conditions are altered levels of insulin signaling proteins such as IR-ß, IRS-1, PI3K, Akt, Glut4 and PGC-1α that hamper insulin-mediated glucose uptake as well as other functions of insulin in the cardiomyocytes and the endothelial cells of the heart. Reduced AMPK, PFK-2 and elevated levels of NADP(H)-dependent oxidases produced by activated M1 macrophages of the adipose tissue and elevated levels of circulating angiotensin are also cause of CVD in diabetes mellitus condition. Insulin sensitizers, angiotensin blockers, superoxide scavengers are used as therapeutics in the amelioration of CVD. It evidently becomes important to unravel the mechanisms of the association between IR and CVDs in order to formulate novel efficient drugs to treat patients suffering from insulin resistance-mediated cardiovascular diseases. The possible associations between insulin resistance and cardiovascular diseases are reviewed here.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 2 , Insulin/metabolism , Oxidative Stress , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Humans , Insulin Resistance/physiology , Risk Factors , Signal Transduction/physiology
15.
Am J Physiol Heart Circ Physiol ; 309(4): H692-701, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26092985

ABSTRACT

The recent introduction of novel anticancer therapies, including bevacizumab (BVZ) and sunitinib (SNT), is associated with an increased risk of cardiotoxicity. However, early identification of left ventricular (LV) systolic dysfunction may facilitate dose modification and avoid the development of advanced heart failure. Using a murine model of BVZ- and SNT-mediated cardiotoxicity, we investigated whether cardiac biomarkers and/or tissue velocity imaging (TVI) using echocardiography can detect early changes in cardiac function, before a decrease in LV ejection fraction is identified. A total of 75 wild-type C57Bl/6 male mice were treated with either 0.9% saline, BVZ, or SNT. Serial monitoring of blood pressure, high-sensitivity troponin I, and echocardiographic indexes were performed over a 14-day study period, after which the mice were euthanized for histological and biochemical analyses. Mice treated with either BVZ or SNT developed systemic hypertension as early as day 7, which increased by day 14. Cardiac biomarkers, specifically high-sensitivity troponin I, were not predictive of early LV systolic dysfunction. Although conventional LV ejection fraction values decreased at day 13 in mice treated with either BVZ or SNT, TVI confirmed early LV systolic dysfunction at day 8. Histological and biochemical analysis demonstrated loss of cellular integrity, increased oxidative stress, and increased cardiac apoptosis in mice treated with BVZ or SNT therapy at day 14. In a murine model of BVZ- or SNT-mediated cardiomyopathy, noninvasive assessment by TVI detected early LV systolic dysfunction before alterations in conventional echocardiographic indexes.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , Heart/drug effects , Indoles/adverse effects , Myocardium/metabolism , Pyrroles/adverse effects , Troponin I/blood , Animals , Bevacizumab , Biomarkers/blood , Blood Pressure , Cardiotoxicity/diagnosis , Cardiotoxicity/etiology , Echocardiography , Heart/physiology , Male , Mice , Mice, Inbred C57BL , Sunitinib , Ventricular Function, Left
16.
Cardiovasc Ultrasound ; 13: 18, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25889218

ABSTRACT

BACKGROUND: Cardio-Oncology is an evolving discipline that focuses on the management of cancer patients who develop cardiovascular complications as a result of their treatment. Although the current combination of surgical resection, radiation, and chemotherapy may lead to a cure in cancer patients, the administration of anti-cancer drugs, in particular Doxorubicin (DOX) and Trastuzumab (TRZ), is associated with an increased risk of cardiotoxicity. Little is known on the potential cardioprotective role of renin angiotensin system (RAS) antagonists in the prevention of DOX+TRZ mediated cardiotoxicity. OBJECTIVE: The aim of the study was to determine whether RAS antagonists would be useful in attenuating DOX+TRZ induced cardiotoxicity. METHODS: A total of 240 C57Bl/6 mice were randomized to prophylactic treatment with placebo, Aliskiren, Perindopril, or Valsartan for a total of 13 weeks. Within each arm, mice received treatment with either DOX, TRZ, or the combination of both drugs. Serial murine echocardiography was performed weekly to characterize the degree of cardiovascular remodeling within each group. RESULTS: In wild-type (WT) mice treated with DOX+TRZ, LV end diastolic internal diameter (LVID) increased from 3.1 ± 0.2 mm at baseline to 4.6 ± 0.3 mm at week 13 (p < 0.05) and the LV fractional shortening (FS) decreased from 52 ± 2% at baseline to 26 ± 2% at week 13 (p < 0.05). Prophylactic treatment with Aliskiren, Perindopril, or Valsartan attenuated the degree of LV cavity dilatation with LVID dimensions of 3.9 ± 0.2 mm, 4.1 ± 0.2 mm, and 4.2 ± 0.1 mm at week 13, respectively (p < 0.05). Similarly, prophylactic treatment with Aliskiren, Perindopril, or Valsartan was partially cardioprotective with FS of 40 ± 1%, 32 ± 1%, and 33 ± 2% at week 13, respectively (p < 0.05). As compared to WT mice receiving DOX+TRZ, prophylactic treatment with RAS inhibition was also associated with improved survival, corroborating the echocardiographic findings. CONCLUSION: The cardiotoxic effects of DOX+TRZ were partially attenuated by the prophylactic administration of RAS antagonists in a chronic murine model of chemotherapy induced cardiac dysfunction.


Subject(s)
Angiotensin Receptor Antagonists/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Doxorubicin/adverse effects , Trastuzumab/adverse effects , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/prevention & control , Animals , Antineoplastic Agents/administration & dosage , Male , Mice , Mice, Inbred C57BL , Renin/antagonists & inhibitors , Renin-Angiotensin System/drug effects , Treatment Outcome , Ultrasonography , Ventricular Dysfunction, Left/diagnostic imaging
17.
Circulation ; 128(11 Suppl 1): S69-78, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-24030423

ABSTRACT

BACKGROUND: Allogeneic mesenchymal stem cells (MSCs) were immunoprivileged early after cardiac implantation and improved heart function in preclinical and clinical studies. However, long-term preclinical studies demonstrated that allogeneic MSCs lost their immunoprivilege and were rejected in the injured myocardium, resulting in recurrent ventricular dysfunction. This study identifies some of the mechanisms responsible for the immune switch in MSCs and suggests a new treatment to maintain immunoprivilege and preserve heart function. METHODS AND RESULTS: Rat MSC immunoprivilege was mediated by prostaglandin E2 (PGE2)-induced secretion of 2 critical chemokines, CCL12 and CCL5. These chemokines stimulated the chemoattraction of T cells toward MSCs, suppressed cytotoxic T-cell proliferation, and induced the production of T regulatory cells. MSCs treated with 5-azacytidine for 24 hours differentiated into myogenic cells after 2 weeks, which was associated with decreased PGE2 and chemokine production and the loss of immunoprivilege. Treatment of differentiated MSCs with PGE2 restored chemokine levels and preserved MSC immunoprivilege. In a rat myocardial infarction model, allogeneic MSCs (3 × 10(6) cells/rat) were injected into the infarct region with or without a biodegradable hydrogel that slowly released PGE2. Five weeks later, the transplanted MSCs expressed myogenic lineage markers and were rejected in the control group, but in the PGE2-treated group, the transplanted cells survived and heart function improved. CONCLUSIONS: Allogeneic MSCs maintained immunoprivilege by PGE2-induced secretion of chemokines CCL12 and CCL5. Differentiation of MSCs decreased PGE2 levels, and immunoprivilege was lost. Maintaining PGE2 levels preserved immunoprivilege after differentiation, prevented rejection of implanted MSCs, and restored cardiac function.


Subject(s)
Dinoprostone/metabolism , Graft Rejection/prevention & control , Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Ventricular Function/physiology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Coculture Techniques , Dinoprostone/physiology , Dinoprostone/therapeutic use , Female , Graft Rejection/metabolism , Graft Rejection/physiopathology , Male , Myocardial Infarction/pathology , Random Allocation , Rats , Rats, Inbred Lew , Rats, Wistar , Transplantation, Homologous , Ventricular Function/drug effects
18.
Can J Physiol Pharmacol ; 92(7): 546-50, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24959994

ABSTRACT

Although breast cancer is one of the leading causes of death in women worldwide, there is an overall improvement in the survival of this patient population. This is likely due to a combination of early detection through screening and awareness, improved targeted biological therapy, and an overall improvement in disease management. Despite the beneficial effects of the 2 anti-cancer drugs doxorubicin (DOX) and trastuzumab (TRZ) in women with breast cancer, development of cardiotoxicity is a major concern. The occurrence of left ventricular systolic dysfunction is unacceptably high in nearly 1 in 4 women treated with DOX+TRZ in the breast cancer setting. In this review, we explore the use of non-invasive cardiac imaging for the early detection of chemotherapy-mediated cardiotoxicity in women with breast cancer, in the hope of preventing end-stage heart disease in this cancer population.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , Breast Neoplasms/drug therapy , Cardiotoxicity/diagnosis , Doxorubicin/adverse effects , Heart/physiopathology , Cardiac Imaging Techniques , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , Early Diagnosis , Female , Humans , Trastuzumab , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/diagnosis , Ventricular Dysfunction, Left/prevention & control
19.
Mol Med Rep ; 29(5)2024 May.
Article in English | MEDLINE | ID: mdl-38488036

ABSTRACT

Doxorubicin (Dox) exhibits a high efficacy in the treatment of numerous types of cancer. However, the beneficial cytotoxic effects of Dox are often accompanied by an increase in the risk of cardiotoxicity. Oxidative stress (OS) plays a key role in Dox­induced cardiomyopathy (DIC). OS in cardiomyocytes disrupts endoplasmic reticulum (ER) function, leading to the accumulation of misfolded/unfolded proteins known as ER stress. ER stress acts as an adaptive mechanism; however, prolonged ER stress together with OS may lead to the initiation of cardiomyocyte apoptosis. The present study aimed to explore the potential of an anti­diabetic drug, empagliflozin (EMPA), in mitigating Dox­induced ER stress and cardiomyocyte apoptosis. In the present study, the effects of 1 h pretreatment of EMPA on Dox­treated cardiomyocytes isolated from Sprague­Dawley rats were investigated. After 24 h, EMPA pre­treatment promoted cell survival in the EMPA + Dox group compared with the Dox group. Results of the present study also demonstrated that EMPA mitigated overall ER stress, as the increased expression of ER stress markers was reduced in the EMPA + Dox group. Additionally, OS, inflammation and expression of ER stress apoptotic proteins were also significantly reduced following EMPA pre­treatment in the EMPA + Dox group. Thus, EMPA may exert beneficial effects on Dox­induced ER stress and may exhibit potential changes that can be utilised to further evaluate the role of EMPA in mitigating DIC.


Subject(s)
Benzhydryl Compounds , Cardiomyopathies , Glucosides , Rats , Animals , Rats, Sprague-Dawley , Cardiomyopathies/metabolism , Myocytes, Cardiac/metabolism , Cardiotoxicity/drug therapy , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Doxorubicin/adverse effects , Apoptosis , Oxidative Stress , Endoplasmic Reticulum Stress
20.
CJC Open ; 6(7): 925-937, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39026621

ABSTRACT

Background: Although the current combination of surgery, radiation, and chemotherapy is used in the breast-cancer setting, the administration of the anticancer drugs doxorubicin and trastuzumab is associated with an increased risk of developing heart failure. The aim of this study is to determine whether dietary flaxseed is comparable and/or synergistic with the angiotensin-converting enzyme inhibitor perindopril in the treatment of doxorubicin- and trastuzumab-mediated cardiotoxicity. Methods: In a chronic in vivo murine model (n = 110), doxorubicin and trastuzumab (8 mg/kg and 3 mg/kg, respectively) were administered weekly for 3 weeks. Following this period, the mice were randomized to daily consumption of a 10% flaxseed supplemented diet, administration of perindopril (3 mg/kg) via oral gavage, or a combination of both flaxseed and perindopril for an additional 3 weeks. Results: In mice treated with doxorubicin and trastuzumab, the left ventricular ejection fraction decreased from 74% ± 4% at baseline to 30% ± 2% at week 6. Treatment with either flaxseed or perindopril, or with flaxseed and perindopril improved left ventricular ejection fraction to 52% ± 4%, 54% ± 4%, and 55% ± 3%, respectively (P < 0.05). Although histologic analyses confirmed significant loss of sarcomere integrity and vacuolization in the doxorubicin- and trastuzumab-treated mice, treatment with flaxseed or perindopril, or with flaxseed and perindopril improved myocyte integrity. Finally, the level of Bcl-2 interacting protein 3, high-mobility group box 1 protein expression, and the levels of select oxylipins, were significantly elevated in mice receiving doxorubicin and trastuzumab; these markers were attenuated by treatment with either flaxseed or perindopril, or with flaxseed and perindopril. Conclusions: Flaxseed was equivalent to perindopril at improving cardiovascular remodelling by reducing biomarkers of inflammation, mitochondrial damage, and cell death.


Contexte: Si l'association actuelle de la chirurgie, de la radiothérapie et de la chimiothérapie est utilisée pour le traitement du cancer du sein, on observe néanmoins que l'administration de la doxorubicine et du trastuzumab, deux anticancéreux, augmente les risques d'insuffisance cardiaque. Cette étude vise à déterminer si les graines de lin alimentaires ont un effet comparable et/ou synergique à celui du périndopril, un inhibiteur de l'enzyme de conversion de l'angiotensine, dans le traitement de la cardiotoxicité liée à la doxorubicine et au trastuzumab. Méthodologie: Dans un modèle murin chronique in vivo (n = 110), la doxorubicine et le trastuzumab (8 mg/kg et 3 mg/kg, respectivement) ont été administrés une fois par semaine pendant trois semaines. Après cette période, les souris ont été réparties de façon aléatoire dans trois groupes : l'un recevant tous les jours un régime alimentaire contenant 10 % de graines de lin, un autre recevant du périndopril (3 mg/kg) par gavage oral et un troisième recevant à la fois des graines de lin et du périndopril pendant trois semaines supplémentaires. Résultats: Chez les souris recevant la doxorubicine et le trastuzumab, la fraction d'éjection ventriculaire gauche est passée de 74 % ± 4 % au départ à 30 % ± 2 % à la semaine 6. Avec le traitement par les graines de lin seules, le périndopril seul ou les graines de lin et le périndopril en association, la fraction d'éjection ventriculaire gauche est passée à 52 % ± 4 %, à 54 % ± 4 % et à 55 % ± 3 %, respectivement (p < 0,05). Bien que les analyses histologiques aient permis de confirmer une perte significative de l'intégrité des sarcomères et une vacuolisation chez les souris recevant la doxorubicine et le trastuzumab, le traitement par les graines de lin seules, le périndopril seul ou les graines de lin et le périndopril en association a amélioré l'intégrité des myocytes. Enfin, les taux de protéine 3 interagissant avec BCL-2, l'expression de la protéine HMGB1 (high-mobility group box 1) et les taux de certaines oxylipines étaient significativement élevés chez les souris recevant la doxorubicine et le trastuzumab. Ces marqueurs ont été atténués par les graines de lin, le périndopril ou l'association des deux. Conclusions: En diminuant les biomarqueurs de l'inflammation, les dommages aux mitochondries et la mort cellulaire, les graines de lin ont un effet équivalant à celui du périndopril quant à l'amélioration du remodelage cardiovasculaire.

SELECTION OF CITATIONS
SEARCH DETAIL