Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 323(1): H176-H200, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35657616

ABSTRACT

Diabetes is a major risk factor for cardiovascular diseases, including diabetic cardiomyopathy, atherosclerosis, myocardial infarction, and heart failure. As cardiovascular disease represents the number one cause of death in people with diabetes, there has been a major emphasis on understanding the mechanisms by which diabetes promotes cardiovascular disease, and how antidiabetic therapies impact diabetic heart disease. With a wide array of models to study diabetes (both type 1 and type 2), the field has made major progress in answering these questions. However, each model has its own inherent limitations. Therefore, the purpose of this guidelines document is to provide the field with information on which aspects of cardiovascular disease in the human diabetic population are most accurately reproduced by the available models. This review aims to emphasize the advantages and disadvantages of each model, and to highlight the practical challenges and technical considerations involved. We will review the preclinical animal models of diabetes (based on their method of induction), appraise models of diabetes-related atherosclerosis and heart failure, and discuss in vitro models of diabetic heart disease. These guidelines will allow researchers to select the appropriate model of diabetic heart disease, depending on the specific research question being addressed.


Subject(s)
Atherosclerosis , Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Heart Failure , Myocardial Infarction , Animals , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/complications , Heart Failure/etiology , Humans , Hypoglycemic Agents , Myocardial Infarction/complications
2.
Antioxidants (Basel) ; 11(1)2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35052647

ABSTRACT

The present study aimed to examine the effects of low doses of angiotensin II (AngII) on cardiac function, myocardial substrate utilization, energetics, and mitochondrial function in C57Bl/6J mice and in a transgenic mouse model with cardiomyocyte specific upregulation of NOX2 (csNOX2 TG). Mice were treated with saline (sham), 50 or 400 ng/kg/min of AngII (AngII50 and AngII400) for two weeks. In vivo blood pressure and cardiac function were measured using plethysmography and echocardiography, respectively. Ex vivo cardiac function, mechanical efficiency, and myocardial substrate utilization were assessed in isolated perfused working hearts, and mitochondrial function was measured in left ventricular homogenates. AngII50 caused reduced mechanical efficiency despite having no effect on cardiac hypertrophy, function, or substrate utilization. AngII400 slightly increased systemic blood pressure and induced cardiac hypertrophy with no effect on cardiac function, efficiency, or substrate utilization. In csNOX2 TG mice, AngII400 induced cardiac hypertrophy and in vivo cardiac dysfunction. This was associated with a switch towards increased myocardial glucose oxidation and impaired mitochondrial oxygen consumption rates. Low doses of AngII may transiently impair cardiac efficiency, preceding the development of hypertrophy induced at higher doses. NOX2 overexpression exacerbates the AngII -induced pathology, with cardiac dysfunction and myocardial metabolic remodelling.

3.
EMBO J ; 39(19): e103530, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33001475

ABSTRACT

Cells subjected to environmental stresses undergo regulated cell death (RCD) when homeostatic programs fail to maintain viability. A major mechanism of RCD is the excessive calcium loading of mitochondria and consequent triggering of the mitochondrial permeability transition (mPT), which is especially important in post-mitotic cells such as cardiomyocytes and neurons. Here, we show that stress-induced upregulation of the ROS-generating protein Nox4 at the ER-mitochondria contact sites (MAMs) is a pro-survival mechanism that inhibits calcium transfer through InsP3 receptors (InsP3 R). Nox4 mediates redox signaling at the MAM of stressed cells to augment Akt-dependent phosphorylation of InsP3 R, thereby inhibiting calcium flux and mPT-dependent necrosis. In hearts subjected to ischemia-reperfusion, Nox4 limits infarct size through this mechanism. These results uncover a hitherto unrecognized stress pathway, whereby a ROS-generating protein mediates pro-survival effects through spatially confined signaling at the MAM to regulate ER to mitochondria calcium flux and triggering of the mPT.


Subject(s)
Calcium Signaling , Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , NADPH Oxidase 4/metabolism , Animals , Cell Survival , Inositol 1,4,5-Trisphosphate Receptors/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , NADPH Oxidase 4/genetics , Oxidative Stress , Rats
4.
Am J Physiol Heart Circ Physiol ; 319(3): H682-H693, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32795177

ABSTRACT

An ischemic insult is accompanied by an acute increase in circulating fatty acid (FA) levels, which can induce adverse changes related to cardiac metabolism/energetics. Although chronic hyperlipidemia contributes to the pathogenesis of obesity-/diabetes-related cardiomyopathy, it is unclear how these hearts are affected by an acute high FA-load. We hypothesize that adaptation to chronic FA exposure enhances the obese hearts' ability to handle an acute high FA-load. Diet-induced obese (DIO) and age-matched control (CON) mouse hearts were perfused in the presence of low- or high FA-load (0.4 and 1.8 mM, respectively). Left ventricular (LV) function, FA oxidation rate, myocardial oxygen consumption, and mechanical efficiency were assessed, followed by analysis of myocardial oxidative stress, mitochondrial respiration, protein acetylation, and gene expression. Finally, ischemic tolerance was determined by examining LV functional recovery and infarct size. Under low-FA conditions, DIO hearts showed mild LV dysfunction, oxygen wasting, mechanical inefficiency, and reduced mitochondrial OxPhos. High FA-load increased FA oxidation rates in both groups, but this did not alter any of the above parameters in DIO hearts. In contrast, CON hearts showed FA-induced mechanical inefficiency, oxidative stress, and reduced OxPhos, as well as enhanced acetylation and activation of PPARα-dependent gene expression. While high FA-load did not alter functional recovery and infarct size in CON hearts, it increased ischemic tolerance in DIO hearts. Thus, this study demonstrates that acute FA-load affects normal and obese hearts differently and that chronically elevated circulating FA levels render the DIO heart less vulnerable to the disadvantageous effects of an acute FA-load.NEW & NOTEWORTHY An acute myocardial fat-load leads to oxidative stress, oxygen wasting, mechanical inefficiency, hyperacetylation, and impaired mitochondrial function, which can contribute to reduced ischemic tolerance. Following obesity/insulin resistance, hearts were less affected by a high fat-load, which subsequently also improved ischemic tolerance. This study highlights that an acute fat-load affects normal and obese hearts differently and that obesity renders hearts less vulnerable to the disadvantageous effects of an acute fat-load.


Subject(s)
Cardiomyopathies/metabolism , Diet, High-Fat , Energy Metabolism , Fatty Acids/metabolism , Myocardial Ischemia/metabolism , Myocardium/metabolism , Obesity/metabolism , Adaptation, Physiological , Animals , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Disease Models, Animal , Male , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardial Ischemia/etiology , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardium/pathology , Obesity/etiology , Obesity/pathology , Obesity/physiopathology , Oxygen Consumption , Reactive Oxygen Species/metabolism , Ventricular Function, Left
5.
Antioxidants (Basel) ; 9(2)2020 Feb 19.
Article in English | MEDLINE | ID: mdl-32093119

ABSTRACT

Obesity and diabetes are independent risk factors for cardiovascular diseases, and they are associated with the development of a specific cardiomyopathy with elevated myocardial oxygen consumption (MVO2) and impaired cardiac efficiency. Although the pathophysiology of this cardiomyopathy is multifactorial and complex, reactive oxygen species (ROS) may play an important role. One of the major ROS-generating enzymes in the cardiomyocytes is nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2), and many potential systemic activators of NOX2 are elevated in obesity and diabetes. We hypothesized that NOX2 activity would influence cardiac energetics and/or the progression of ventricular dysfunction following obesity. Myocardial ROS content and mechanoenergetics were measured in the hearts from diet-induced-obese wild type (DIOWT) and global NOK2 knock-out mice (DIOKO) and in diet-induced obese C57BL/6J mice given normal water (DIO) or water supplemented with the NOX2-inhibitor apocynin (DIOAPO). Mitochondrial function and ROS production were also assessed in DIO and DIOAPO mice. This study demonstrated that ablation and pharmacological inhibition of NOX2 both improved mechanical efficiency and reduced MVO2 for non-mechanical cardiac work. Mitochondrial ROS production was also reduced following NOX2 inhibition, while cardiac mitochondrial function was not markedly altered by apocynin-treatment. Therefore, these results indicate a link between obesity-induced myocardial oxygen wasting, NOX2 activation, and mitochondrial ROS.

6.
Front Physiol ; 10: 1274, 2019.
Article in English | MEDLINE | ID: mdl-31632301

ABSTRACT

Physical activity is an efficient strategy to delay development of obesity and insulin resistance, and thus the progression of obesity/diabetes-related cardiomyopathy. In support of this, experimental studies using animal models of obesity show that chronic exercise prevents the development of obesity-induced cardiac dysfunction (cardiomyopathy). Whether exercise also improves the tolerance to ischemia-reperfusion in these models is less clear, and may depend on the type of exercise procedure as well as time of initiation. We have previously shown a reduction in ischemic-injury in diet-induced obese mice, when the exercise was started prior to the development of cardiac dysfunction in this model. In the present study, we aimed to explore the effect of exercise on ischemic-tolerance when exercise was initiated after the development obesity-mediated. Male C57BL/6J mice were fed a high-fat diet (HFD) for 20-22 weeks, where they were subjected to high-intensity interval training (HIT) during the last 3 weeks of the feeding period. Sedentary HFD fed and chow fed mice served as controls. Left-ventricular (LV) post-ischemic functional recovery and infarct size were measured in isolated perfused hearts. We also assessed the effect of 3-week HIT on mitochondrial function and myocardial oxygen consumption (MVO2). Sedentary HFD fed mice developed marked obesity and insulin resistance, and demonstrated reduced post-ischemic cardiac functional recovery and increased infarct size. Three weeks of HIT did not induce cardiac hypertrophy and only had a mild effect on obesity and insulin resistance. Despite this, HIT improved post-ischemic LV functional recovery and reduced infarct size. This increase in ischemic-tolerance was accompanied by an improved mitochondrial function as well as reduced MVO2. The present study highlights the beneficial effects of exercise training with regard to improving the ischemic-tolerance in hearts with cardiomyopathy following obesity and insulin resistance. This study also emphasizes the exercise-induced improvement of cardiac energetics and mitochondrial function in obesity/diabetes.

7.
Elife ; 72018 12 27.
Article in English | MEDLINE | ID: mdl-30589411

ABSTRACT

Regular exercise has widespread health benefits. Fundamental to these beneficial effects is the ability of the heart to intermittently and substantially increase its performance without incurring damage, but the underlying homeostatic mechanisms are unclear. We identify the ROS-generating NADPH oxidase-4 (Nox4) as an essential regulator of exercise performance in mice. Myocardial Nox4 levels increase during acute exercise and trigger activation of the transcription factor Nrf2, with the induction of multiple endogenous antioxidants. Cardiomyocyte-specific Nox4-deficient (csNox4KO) mice display a loss of exercise-induced Nrf2 activation, cardiac oxidative stress and reduced exercise performance. Cardiomyocyte-specific Nrf2-deficient (csNrf2KO) mice exhibit similar compromised exercise capacity, with mitochondrial and cardiac dysfunction. Supplementation with an Nrf2 activator or a mitochondria-targeted antioxidant effectively restores cardiac performance and exercise capacity in csNox4KO and csNrf2KO mice respectively. The Nox4/Nrf2 axis therefore drives a hormetic response that is required for optimal cardiac mitochondrial and contractile function during physiological exercise.


Subject(s)
Myocardium/enzymology , NADPH Oxidase 4/metabolism , Physical Conditioning, Animal/physiology , Physiological Phenomena/physiology , Animals , Antioxidants/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Myocardium/metabolism , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , NADPH Oxidase 4/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Signal Transduction
8.
PLoS One ; 13(10): e0204843, 2018.
Article in English | MEDLINE | ID: mdl-30273374

ABSTRACT

Although murine models for studying the development of cardiac dysfunction in diabetes mellitus are well established, their reported cardiac phenotypes vary. These reported divergences may, in addition to the severity of different models, also be linked to the methods used for cardiac functional assessment. In the present study, we examined the functional changes using conventional transthoracic echocardiography (in vivo) and isolated heart perfusion techniques (ex vivo), in hearts from two mouse models; one with an overt type 2 diabetes (the db/db mouse) and one with a prediabetic state, where obesity was induced by a high-fat diet (HFD). Analysis of left ventricular function in the isolated working hearts from HFD-fed mice, suggested that these hearts develop diastolic dysfunction with preserved systolic function. Accordingly, in vivo examination demonstrated maintained systolic function, but we did not find parameters of diastolic function to be altered. In db/db mice, ex vivo working hearts showed both diastolic and systolic dysfunction. Although in vivo functional assessment revealed signs of diastolic dysfunction, the hearts did not display reduced systolic function. The contrasting results between ex vivo and in vivo function could be due to systemic changes that may sustain in vivo function, or a lack of sensitivity using conventional transthoracic echocardiography. Thus, this study demonstrates that the isolated perfused working heart preparation provides unique additional information related to the development of cardiomyopathy, which might otherwise go unnoticed when only using conventional echocardiographic assessment.


Subject(s)
Cardiomyopathies/diagnosis , Diabetes Mellitus, Type 2/complications , Diet, High-Fat/adverse effects , Isolated Heart Preparation/methods , Prediabetic State/complications , Animals , Cardiomyopathies/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Echocardiography , Heart/physiopathology , Male , Mice , Phenotype , Prediabetic State/chemically induced , Prediabetic State/physiopathology , Sensitivity and Specificity
9.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt B): 1908-1913, 2018 May.
Article in English | MEDLINE | ID: mdl-28754449

ABSTRACT

Systemic changes during diabetes such as high glucose, dyslipidemia, hormonal changes and low grade inflammation, are believed to induce structural and functional changes in the cardiomyocyte associated with the development of diabetic cardiomyopathy. One of the hallmarks of the diabetic heart is increased oxidative stress. NADPH-oxidases (NOXs) are important ROS-producing enzymes in the cardiomyocyte mediating both adaptive and maladaptive changes in the heart. NOXs have been suggested as a therapeutic target for several diabetic complications, but their role in diabetic cardiomyopathy is far from elucidated. In this review we aim to provide an overview of the current knowledge regarding the understanding of how NOXs influences cardiac adaptive and maladaptive processes in a "diabetic milieu". This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.


Subject(s)
Diabetes Mellitus/enzymology , Diabetic Cardiomyopathies/enzymology , Myocytes, Cardiac/enzymology , NADPH Oxidases/metabolism , Adaptation, Physiological , Animals , Blood Glucose/metabolism , Calcium/metabolism , Calcium Signaling , Diabetes Mellitus/pathology , Diabetes Mellitus/physiopathology , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Energy Metabolism , Heart/physiopathology , Humans , Insulin Resistance , Lipids/blood , Myocytes, Cardiac/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism
10.
JCI Insight ; 2(24)2017 12 21.
Article in English | MEDLINE | ID: mdl-29263294

ABSTRACT

Cardiac hypertrophic remodeling during chronic hemodynamic stress is associated with a switch in preferred energy substrate from fatty acids to glucose, usually considered to be energetically favorable. The mechanistic interrelationship between altered energy metabolism, remodeling, and function remains unclear. The ROS-generating NADPH oxidase-4 (Nox4) is upregulated in the overloaded heart, where it ameliorates adverse remodeling. Here, we show that Nox4 redirects glucose metabolism away from oxidation but increases fatty acid oxidation, thereby maintaining cardiac energetics during acute or chronic stresses. The changes in glucose and fatty acid metabolism are interlinked via a Nox4-ATF4-dependent increase in the hexosamine biosynthetic pathway, which mediates the attachment of O-linked N-acetylglucosamine (O-GlcNAcylation) to the fatty acid transporter CD36 and enhances fatty acid utilization. These data uncover a potentially novel redox pathway that regulates protein O-GlcNAcylation and reprograms cardiac substrate metabolism to favorably modify adaptation to chronic stress. Our results also suggest that increased fatty acid oxidation in the chronically stressed heart may be beneficial.


Subject(s)
Acetylglucosamine/metabolism , Cardiomegaly/physiopathology , Myocardium/metabolism , NADPH Oxidase 4/physiology , Stress, Physiological/physiology , Adaptation, Physiological/physiology , Animals , Cardiomegaly/metabolism , Energy Metabolism/physiology , Fatty Acids/metabolism , Glucose/metabolism , Glycolysis/physiology , Hexosamines/biosynthesis , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/metabolism , NADPH Oxidase 4/deficiency , NADPH Oxidase 4/genetics , Oxidation-Reduction , Proteome/metabolism
11.
Am J Physiol Heart Circ Physiol ; 313(5): H1054-H1062, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28801525

ABSTRACT

Exercise training is a potent therapeutic approach in obesity and diabetes that exerts protective effects against the development of diabetic cardiomyopathy and ischemic injury. Acute increases in circulating fatty acids (FAs) during an ischemic insult can challenge the heart, since high FA load is considered to have adverse cardiac effects. In the present study, we tested the hypothesis that exercise-induced cardiac effects in diet-induced obese mice are abrogated by an acute high FA load. Diet-induced obese mice were fed a high-fat diet (HFD) for 20 wk. They were exercised using moderate- and/or high-intensity exercise training (MIT and HIT, respectively) for 10 or 3 wk, and isolated perfused hearts from these mice were exposed to a high FA load. Sedentary HFD mice served as controls. Ventricular function and myocardial O2 consumption were assessed after 10 wk of HIT and MIT, and postischemic functional recovery and infarct size were examined after 3 wk of HIT. In addition to improving aerobic capacity and reducing obesity and insulin resistance, long-term exercise ameliorated the development of diet-induced cardiac dysfunction. This was associated with improved mechanical efficiency because of reduced myocardial oxygen consumption. Although to a lesser extent, 3-wk HIT also increased aerobic capacity and decreased obesity and insulin resistance. HIT also improved postischemic functional recovery and reduced infarct size. Event upon the exposure to a high FA load, short-term exercise induced an oxygen-sparing effect. This study therefore shows that exercise-induced cardioprotective effects are present under hyperlipidemic conditions and highlights the important role of myocardial energetics during ischemic stress.NEW & NOTEWORTHY The exercise-induced cardioprotective effects in obese hearts are present under hyperlipidemic conditions, comparable to circulating levels of FA occurring with an ischemic insult. Myocardial oxygen sparing is associated with this effect, despite the general notion that high fat can decrease cardiac efficiency. This highlights the role of myocardial energetics during ischemic stress.


Subject(s)
Diet, High-Fat , Energy Metabolism , Exercise Therapy , Fatty Acids/metabolism , Myocardial Infarction/prevention & control , Myocardium/metabolism , Obesity/therapy , Oxygen Consumption , Animals , Blood Glucose/metabolism , Disease Models, Animal , Exercise Tolerance , Insulin Resistance , Isolated Heart Preparation , Male , Mice, Inbred C57BL , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/pathology , Obesity/etiology , Obesity/metabolism , Obesity/physiopathology , Recovery of Function , Time Factors , Ventricular Function, Left
12.
EMBO J ; 35(3): 319-34, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26742780

ABSTRACT

Phosphorylation of translation initiation factor 2α (eIF2α) attenuates global protein synthesis but enhances translation of activating transcription factor 4 (ATF4) and is a crucial evolutionarily conserved adaptive pathway during cellular stresses. The serine-threonine protein phosphatase 1 (PP1) deactivates this pathway whereas prolonging eIF2α phosphorylation enhances cell survival. Here, we show that the reactive oxygen species-generating NADPH oxidase-4 (Nox4) is induced downstream of ATF4, binds to a PP1-targeting subunit GADD34 at the endoplasmic reticulum, and inhibits PP1 activity to increase eIF2α phosphorylation and ATF4 levels. Other PP1 targets distant from the endoplasmic reticulum are unaffected, indicating a spatially confined inhibition of the phosphatase. PP1 inhibition involves metal center oxidation rather than the thiol oxidation that underlies redox inhibition of protein tyrosine phosphatases. We show that this Nox4-regulated pathway robustly enhances cell survival and has a physiologic role in heart ischemia-reperfusion and acute kidney injury. This work uncovers a novel redox signaling pathway, involving Nox4-GADD34 interaction and a targeted oxidative inactivation of the PP1 metal center, that sustains eIF2α phosphorylation to protect tissues under stress.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , NADPH Oxidases/metabolism , Protein Phosphatase 1/metabolism , Receptors, Neuropeptide Y/antagonists & inhibitors , Signal Transduction , Animals , Cell Line , Humans , NADPH Oxidase 4 , Oxidation-Reduction
13.
PLoS One ; 10(11): e0143095, 2015.
Article in English | MEDLINE | ID: mdl-26566220

ABSTRACT

OBJECTIVE: To investigate the effects of high intensity interval training (HIIT) on the maternal heart, fetuses and placentas of pregnant rats. METHODS: Female Sprague-Dawley rats were randomly assigned to HIIT or sedentary control groups. The HIIT group was trained for 6 weeks with 10 bouts of high intensity uphill running on a treadmill for four minutes (at 85-90% of maximal oxygen consumption) for five days/week. After three weeks of HIIT, rats were mated. After six weeks (gestational day 20 in pregnant rats), echocardiography was performed to evaluate maternal cardiac function. Real-time PCR was performed for the quantification of gene expression, and oxidative stress and total antioxidant capacity was assessed in the tissue samples. RESULTS: Maternal heart weight and systolic function were not affected by HIIT or pregnancy. In the maternal heart, expression of 11 of 22 genes related to cardiac remodeling was influenced by pregnancy but none by HIIT. Litter size, fetal weight and placental weight were not affected by HIIT. Total antioxidant capacity, malondialdehyde content, peroxidase and superoxide dismutase activity measured in the placenta, fetal heart and liver were not influenced by HIIT. HIIT reduced the expression of eNOS (p = 0.03), hypoxia-inducible factor 1α (p = 0.04) and glutathione peroxidase 4.2 (p = 0.02) in the fetal liver and increased the expression of vascular endothelial growth factor-ß (p = 0.014), superoxide dismutase 1 (p = 0.001) and tissue inhibitor of metallopeptidase 3 (p = 0.049) in the fetal heart. CONCLUSIONS: Maternal cardiac function and gene expression was not affected by HIIT. Although HIIT did not affect fetal growth, level of oxidative stress and total antioxidant capacity in the fetal tissues, some genes related to oxidative stress were altered in the fetal heart and liver indicating that protective mechanisms may be activated.


Subject(s)
Heart/embryology , Liver/embryology , Physical Conditioning, Animal , Placenta/embryology , Pregnancy, Animal , Animals , Antioxidants/chemistry , Exercise Test , Female , Gene Expression Profiling , Glutathione Peroxidase/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Malondialdehyde/chemistry , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Oxygen Consumption , Phospholipid Hydroperoxide Glutathione Peroxidase , Pregnancy , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Tissue Inhibitor of Metalloproteinase-3/metabolism , Vascular Endothelial Growth Factor B/metabolism
14.
Lancet ; 385 Suppl 1: S73, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-26312895

ABSTRACT

BACKGROUND: The mechanisms that determine whether the heart adapts to overload stress, or fails, are poorly understood. NADPH oxidase (NOX) proteins produce reactive oxygen species (ROS) involved in redox signalling, and our recent studies have found that an increase in Nox4 during pressure overload protects the heart against failure. We aimed to identify novel Nox4-driven cardioprotective mechanisms that promote adaptive cardiac remodelling. METHODS: We first undertook a proteomic comparison of heart tissue from cardiac-targeted Nox4-overexpressing mice and controls. The Nox4 cardiac metabolome was then investigated by (1)H nuclear magnetic resonance (NMR) spectroscopy. Effects on cardiac metabolism were assessed by ex-vivo working heart perfusions and isolated mitochondrial respiration studies. Ex-vivo cardiac energetics were assessed by (31)P NMR. Alterations to cardiac fatty acid oxidation were explored in primary cardiomyocytes (extracellular flux analysis). FINDINGS: Cardiac-targeted Nox4 overexpression profoundly remodelled the cardiac proteome in an isoform-specific manner, both in the unstressed and stressed heart. Glycolysis and fatty acid oxidation were identified as the most enriched pathways that were altered by Nox4. Metabolomic analysis showed a 2·2 times increase in acetylcarnitine concentrations (p=0·002). Ex-vivo heart perfusions demonstrated a profound increase in palmitate oxidation relative to wild-type hearts (3·6 times increase, p=0·01), with opposite findings observed in primary cardiomyocytes with a knockdown of Nox4. A preference for fatty acid oxidation in Nox4 hearts correlated with a better energetic state (phosphocreatine:ATP ratio) when subjected to increasing doses of isoprenaline stress under baseline and pressure-overload. INTERPRETATION: In this study we identified a novel role for Nox4 in the regulation of cardiac fatty acid oxidation. Cardiomyocyte-targeted Nox4 hearts preferentially oxidised fatty acids for energy provision, improving myocardial energetics under stress. Enhancing fatty acid oxidation might have an adaptive role in the setting of pressure-overload hypertrophy. These data provide novel insights into ROS-dependent metabolic programming. FUNDING: UK Medical Research Council, British Heart Foundation.

15.
Antioxid Redox Signal ; 22(17): 1587-605, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-25738326

ABSTRACT

SIGNIFICANCE: Over-nutrition and sedentary lifestyle has led to a worldwide increase in obesity, insulin resistance, and type 2 diabetes (T2D) associated with an increased risk of development of cardiovascular disorders. Diabetic cardiomyopathy, independent of hypertension or coronary disease, is induced by a range of systemic changes and may through multiple processes result in functional and structural cardiac derangements. The pathogenesis of this cardiomyopathy is complex and multifactorial, and it will eventually lead to reduced cardiac working capacity and increased susceptibility to ischemic injury. RECENT ADVANCES: Metabolic disturbances such as altered lipid handling and substrate utilization, decreased mechanical efficiency, mitochondrial dysfunction, disturbances in nonoxidative glucose pathways, and increased oxidative stress are hallmarks of diabetic cardiomyopathy. Interestingly, several of these disturbances are found to precede the development of cardiac dysfunction. CRITICAL ISSUES: Exercise training is effective in the prevention and treatment of obesity and T2D. In addition to its beneficial influence on diabetes/obesity-related systemic changes, it may also amend many of the metabolic disturbances characterizing the diabetic myocardium. These changes are due to both indirect effects, exercise-mediated systemic changes, and direct effects originating from the high contractile activity of the heart during physical training. FUTURE DIRECTIONS: Revealing the molecular mechanisms behind the beneficial effects of exercise training is of considerable scientific value to generate evidence-based therapy and in the development of new treatment strategies.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Exercise , Animals , Calcium/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Humans , Lipid Metabolism , Mitochondria/metabolism , Myocardium/metabolism , Myocytes, Cardiac/pathology , Reactive Oxygen Species/metabolism
16.
J Cell Physiol ; 230(1): 160-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24905939

ABSTRACT

Matrix metalloproteinase 2 (MMP-2) is a proteolytic enzyme implicated in motility, differentiation, and regeneration of skeletal muscle fibers through processing of extracellular substrates. Although MMP-2 has been found to be localized intracellularly in cardiomyocytes where the enzyme is thought to contribute to post-ischemic loss of contractility, little is known about intracellular MMP-2 activity in skeletal muscle fibers. In the present study we demonstrate intracellular MMP-2 in normal skeletal muscle by immunohistochemical staining. Immunogold electron microscopic analyses indicated that the enzyme was concentrated in Z-lines of the sarcomers, in the nuclear membrane, and in mitochondria. By use of in situ zymography, we found that gelatinolytic activity in muscle fibers was co-localized with immunofluorecent staining for MMP-2. Staining for MMP-9, the other member of the gelatinase group of the MMPs, was negative. The broad-spectrum metalloprotease inhibitor EDTA and the selective gelatinase inhibitor CTT2, but not the cysteine inhibitor E64, strongly reduced the gelatinolytic activity. The intracellular gelatinolytic activity was much more prominent in fast twitch type II fibers than in slow twitch type I fibers, and there was a decrease in intracellular gelatinolytic activity and MMP-2 expression in muscles from mice exposed to high intensity interval training. Together our results indicate that MMP-2 is part of the intracellular proteolytic network in normal skeletal muscle, especially in fast twitch type II fibers. Further, the results suggest that intracellular MMP-2 in skeletal muscle fibers is active during normal homeostasis, and affected by the level of physical activity.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Muscle Fibers, Fast-Twitch/enzymology , Muscle Fibers, Slow-Twitch/enzymology , Animals , Cell Differentiation , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Edetic Acid/pharmacology , Gelatinases/antagonists & inhibitors , Leucine/analogs & derivatives , Leucine/pharmacology , Male , Matrix Metalloproteinase 2/biosynthesis , Mice , Mice, Inbred C57BL , Muscle Cells/cytology , Myoblasts/cytology , Peptides, Cyclic/pharmacology , Physical Conditioning, Animal , Sarcomeres/metabolism
17.
J Nutr ; 144(2): 164-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24285691

ABSTRACT

We showed previously that dietary supplementation with oil from the marine zooplankton Calanus finmarchicus (Calanus oil) attenuates obesity, inflammation, and glucose intolerance in mice. More than 80% of Calanus oil consists of wax esters, i.e., long-chain fatty alcohols linked to long-chain fatty acids. In the present study, we compared the metabolic effects of Calanus oil-derived wax esters (WE) with those of purified eicosapentaenoic acid (EPA) + docosahexaenoic acid (DHA) ethyl esters (E/D) in a mouse model of diet-induced obesity. C57BL/6J mice received a high-fat diet (HFD; 45% energy from fat). After 7 wk, the diet was supplemented with either 1% (wt:wt) WE or 0.2% (wt:wt) E/D. The amount of EPA + DHA in the E/D diet was matched to the total amount of n-3 (ω-3) polyunsaturated fatty acids (PUFAs) in the WE diet. A third group was given an unsupplemented HFD throughout the entire 27-wk feeding period. WE reduced body weight gain, abdominal fat, and liver triacylglycerol by 21%, 34%, and 52%, respectively, and significantly improved glucose tolerance and aerobic capacity. In abdominal fat depots, WE reduced macrophage infiltration by 74% and downregulated expression of proinflammatory genes (tumor necrosis factor-α, interleukin-6, and monocyte chemoattractant protein-1), whereas adiponectin expression was significantly upregulated. By comparison, E/D primarily suppressed the expression of proinflammatory genes but had less influence on glucose tolerance than WE. E/D affected obesity parameters, aerobic capacity, or adiponectin expression by <10%. These results show that the wax ester component of Calanus oil can account for the biologic effects shown previously for the crude oil. However, these effects cannot exclusively be ascribed to the content of n-3 PUFAs in the wax ester fraction.


Subject(s)
Biological Products/therapeutic use , Copepoda/chemistry , Fatty Acids, Omega-3/therapeutic use , Metabolic Diseases/prevention & control , Obesity/prevention & control , Waxes/therapeutic use , Zooplankton/chemistry , Abdominal Fat/drug effects , Abdominal Fat/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Animals , Biological Products/pharmacology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Diet/adverse effects , Dietary Supplements , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Down-Regulation , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/therapeutic use , Esters/pharmacology , Esters/therapeutic use , Fatty Acids, Omega-3/pharmacology , Gene Expression/drug effects , Glucose Intolerance/etiology , Glucose Intolerance/prevention & control , Inflammation/genetics , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Liver/drug effects , Liver/metabolism , Macrophages/drug effects , Male , Metabolic Diseases/etiology , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Physical Endurance/drug effects , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Waxes/pharmacology , Weight Gain/drug effects
18.
Basic Res Cardiol ; 108(4): 359, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23740217

ABSTRACT

Heart failure and many of the conditions that predispose to heart failure are associated with oxidative stress. This is considered to be important in the pathophysiology of the condition but clinical trials of antioxidant approaches to prevent cardiovascular morbidity and mortality have been unsuccessful. Part of the reason for this may be the failure to appreciate the complexity of the effects of reactive oxygen species. At one extreme, excessive oxidative stress damages membranes, proteins and DNA but lower levels of reactive oxygen species may exert much more subtle and specific regulatory effects (termed redox signalling), even on physiological signalling pathways. In this article, we review our current understanding of the roles of such redox signalling pathways in the pathophysiology of heart failure, including effects on cardiomyocyte hypertrophy signalling, excitation-contraction coupling, arrhythmia, cell viability and energetics. Reactive oxygen species generated by NADPH oxidase proteins appear to be especially important in redox signalling. The delineation of specific redox-sensitive pathways and mechanisms that contribute to different components of the failing heart phenotype may facilitate the development of newer targeted therapies as opposed to the failed general antioxidant approaches of the past.


Subject(s)
Heart Failure/physiopathology , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Animals , Disease Models, Animal , Heart Failure/metabolism , Humans , Oxidation-Reduction , Oxidative Stress/physiology
19.
Br J Nutr ; 110(12): 2186-93, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23768435

ABSTRACT

The aim of the present study was to investigate the effects of oil extracted from the zooplankton Calanus finmarchicus (Calanus oil) on diet-induced obesity and obesity-related disorders in mice. C57BL/6J mice fed a high-fat diet (HFD, 45% energy from fat) exhibited increased body weight and abdominal fat accumulation as well as impaired glucose tolerance compared with mice fed a normal chow diet (10% energy from fat). Supplementing the HFD with 1.5% (w/w) Calanus oil reduced body-weight gain, abdominal fat accumulation and hepatic steatosis by 16, 27 and 41%, respectively, and improved glucose tolerance by 16%. Calanus oil supplementation reduced adipocyte size and increased the mRNA expression of adiponectin in adipose tissue. It also reduced macrophage infiltration by more than 70%, accompanied by reduced mRNA expression of pro-inflammatory cytokines (TNF-α, IL-6 and monocyte chemotactic protein-1). The effects of Calanus oil were not only preventive, but also therapeutic, as the oil proved to be beneficial, regardless of whether the supplementation was started before or after the onset of obesity and glucose intolerance. Although the present study cannot pinpoint the active component(s) of the oil, there is reason to believe that the n-3 fatty acids EPA and DHA and/or antioxidants are responsible for its beneficial effects. It should be noted that the concentration of n-3 fatty acids in the Calanus oil diet was considerably lower than the concentrations used in similar studies reporting beneficial effects on obesity and obesity-related abnormalities.


Subject(s)
Abdominal Fat/drug effects , Biological Products/therapeutic use , Copepoda/chemistry , Glucose Intolerance/drug therapy , Obesity/drug therapy , Weight Gain/drug effects , Zooplankton/chemistry , Abdominal Fat/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Biological Products/pharmacology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat/adverse effects , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/prevention & control , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Obesity/metabolism , RNA, Messenger/metabolism
20.
Diabetes ; 62(7): 2287-94, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23493573

ABSTRACT

Although exercise reduces several cardiovascular risk factors associated with obesity/diabetes, the metabolic effects of exercise on the heart are not well-known. This study was designed to investigate whether high-intensity interval training (HIT) is superior to moderate-intensity training (MIT) in counteracting obesity-induced impairment of left ventricular (LV) mechanoenergetics and function. C57BL/6J mice with diet-induced obesity (DIO mice) displaying a cardiac phenotype with altered substrate utilization and impaired mechanoenergetics were subjected to a sedentary lifestyle or 8-10 weeks of isocaloric HIT or MIT. Although both modes of exercise equally improved aerobic capacity and reduced obesity, only HIT improved glucose tolerance. Hearts from sedentary DIO mice developed concentric LV remodeling with diastolic and systolic dysfunction, which was prevented by both HIT and MIT. Both modes of exercise also normalized LV mechanical efficiency and mechanoenergetics. These changes were associated with altered myocardial substrate utilization and improved mitochondrial capacity and efficiency, as well as reduced oxidative stress, fibrosis, and intracellular matrix metalloproteinase 2 content. As both modes of exercise equally ameliorated the development of diabetic cardiomyopathy by preventing LV remodeling and mechanoenergetic impairment, this study advocates the therapeutic potential of physical activity in obesity-related cardiac disorders.


Subject(s)
Heart Ventricles/physiopathology , Obesity/physiopathology , Physical Conditioning, Animal/physiology , Ventricular Function/physiology , Animals , Diet, High-Fat/adverse effects , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/physiology , Myocardium/metabolism , Obesity/etiology , Obesity/metabolism , Oxidative Stress/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...