101
|
Liu C, Zhang C, Wang W, Yuan F, He T, Chen Y, Wang Q, Huang J. Integrated metabolomics and network toxicology to reveal molecular mechanism of celastrol induced cardiotoxicity. Toxicol Appl Pharmacol 2019; 383:114785. [PMID: 31629732 DOI: 10.1016/j.taap.2019.114785] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/31/2019] [Accepted: 10/15/2019] [Indexed: 12/20/2022]
Abstract
Celastrol (CS), an active triterpene derived from traditional Chinese medicine Tripterygium wilfordii Hook. f, has been used to treat chronic inflammation, arthritis and other diseases. However, it has been reported that CS can trigger cardiotoxicity and the molecular mechanism of heart injury induced by CS is not clear. Considering the wide application of Tripterygium wilfordii Hook. f in clinics, it is necessary to develop an accurate and reliable method to assess the safety of CS, and to elucidate as much as possible the mechanism of cardiotoxicity induced by CS. In this study, Ultra-performance liquid chromatography coupled with quadrupole time of flight mass spectrometry (UPLC-Q-TOF/MS)-based metabolomics revealed clues to the mechanism of CS-induced heart injury. Palmitic acid significantly increased in plasma from CS-treated rats, and this increase resulted in oxidative stress response in vivo. Excessive ROS further activate TNF signaling pathway and caspase family, which were obtained from the KEGG enrichment analysis of network toxicology strategy. Protein expression level of caspase-3, caspase-8, bax were significantly increased by western blot. Q-PCR also showed the similar results as western blot. It means that apoptosis plays a key role in the process of celastrol induced cardiotoxicity. Blocking this signal axis may be a potential way to protect myocardial tissue.
Collapse
Affiliation(s)
- Chuanxin Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing 102488, China
| | - Chenning Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing 102488, China
| | - Wenxin Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing 102488, China
| | - Fuli Yuan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing 102488, China
| | - Tao He
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing 102488, China
| | - Yahong Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing 102488, China
| | - Qiang Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing 102488, China
| | - Jianmei Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing 102488, China..
| |
Collapse
|
102
|
Goldberg IJ, Reue K, Abumrad NA, Bickel PE, Cohen S, Fisher EA, Galis ZS, Granneman JG, Lewandowski ED, Murphy R, Olive M, Schaffer JE, Schwartz-Longacre L, Shulman GI, Walther TC, Chen J. Deciphering the Role of Lipid Droplets in Cardiovascular Disease: A Report From the 2017 National Heart, Lung, and Blood Institute Workshop. Circulation 2019; 138:305-315. [PMID: 30012703 DOI: 10.1161/circulationaha.118.033704] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lipid droplets (LDs) are distinct and dynamic organelles that affect the health of cells and organs. Much progress has been made in understanding how these structures are formed, how they interact with other cellular organelles, how they are used for storage of triacylglycerol in adipose tissue, and how they regulate lipolysis. Our understanding of the biology of LDs in the heart and vascular tissue is relatively primitive in comparison with LDs in adipose tissue and liver. The National Heart, Lung, and Blood Institute convened a working group to discuss how LDs affect cardiovascular diseases. The goal of the working group was to examine the current state of knowledge on the cell biology of LDs, including current methods to study them in cells and organs and reflect on how LDs influence the development and progression of cardiovascular diseases. This review summarizes the working group discussion and recommendations on research areas ripe for future investigation that will likely improve our understanding of atherosclerosis and heart function.
Collapse
Affiliation(s)
| | - Karen Reue
- University of California, Los Angeles (K.R.)
| | | | - Perry E Bickel
- University of Texas Southwestern Medical Center, Dallas (P.E.B.)
| | - Sarah Cohen
- University of North Carolina at Chapel Hill (S.C.)
| | | | - Zorina S Galis
- National Institutes of Health/National, Heart, Lung, and Blood Institute, Bethesda, MD (Z.S.G., M.O., L.S.-L., J.C.)
| | | | | | | | - Michelle Olive
- National Institutes of Health/National, Heart, Lung, and Blood Institute, Bethesda, MD (Z.S.G., M.O., L.S.-L., J.C.)
| | | | - Lisa Schwartz-Longacre
- National Institutes of Health/National, Heart, Lung, and Blood Institute, Bethesda, MD (Z.S.G., M.O., L.S.-L., J.C.)
| | - Gerald I Shulman
- Yale University, Howard Hughes Medical Institute, New Haven, CT (G.I.S.)
| | - Tobias C Walther
- Harvard University, Howard Hughes Medical Institute, Boston, MA (T.C.W.)
| | - Jue Chen
- National Institutes of Health/National, Heart, Lung, and Blood Institute, Bethesda, MD (Z.S.G., M.O., L.S.-L., J.C.).
| |
Collapse
|
103
|
Martinez-Mateu L, Saiz J, Aromolaran AS. Differential Modulation of IK and ICa,L Channels in High-Fat Diet-Induced Obese Guinea Pig Atria. Front Physiol 2019; 10:1212. [PMID: 31607952 PMCID: PMC6773813 DOI: 10.3389/fphys.2019.01212] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022] Open
Abstract
Obesity mechanisms that make atrial tissue vulnerable to arrhythmia are poorly understood. Voltage-dependent potassium (IK, IKur, and IK1) and L-type calcium currents (ICa,L) are electrically relevant and represent key substrates for modulation in obesity. We investigated whether electrical remodeling produced by high-fat diet (HFD) alone or in concert with acute atrial stimulation were different. Electrophysiology was used to assess atrial electrical function after short-term HFD-feeding in guinea pigs. HFD atria displayed spontaneous beats, increased IK (IKr + IKs) and decreased ICa,L densities. Only with pacing did a reduction in IKur and increased IK1 phenotype emerge, leading to a further shortening of action potential duration. Computer modeling studies further indicate that the measured changes in potassium and calcium current densities contribute prominently to shortened atrial action potential duration in human heart. Our data are the first to show that multiple mechanisms (shortened action potential duration, early afterdepolarizations and increased incidence of spontaneous beats) may underlie initiation of supraventricular arrhythmias in obese guinea pig hearts. These results offer different mechanistic insights with implications for obese patients harboring supraventricular arrhythmias.
Collapse
Affiliation(s)
- Laura Martinez-Mateu
- Centro de Investigación e Innovación en Bioingeniería, Universitat Politècnica de València, Valencia, Spain
| | - Javier Saiz
- Centro de Investigación e Innovación en Bioingeniería, Universitat Politècnica de València, Valencia, Spain
| | - Ademuyiwa S Aromolaran
- Cardiac Electrophysiology and Metabolism Research Group, VA New York Harbor Healthcare System, Brooklyn, NY, United States.,Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Physiology & Cellular Biophysics, Columbia University, New York, NY, United States
| |
Collapse
|
104
|
Abstract
Metabolic pathways integrate to support tissue homeostasis and to prompt changes in cell phenotype. In particular, the heart consumes relatively large amounts of substrate not only to regenerate ATP for contraction but also to sustain biosynthetic reactions for replacement of cellular building blocks. Metabolic pathways also control intracellular redox state, and metabolic intermediates and end products provide signals that prompt changes in enzymatic activity and gene expression. Mounting evidence suggests that the changes in cardiac metabolism that occur during development, exercise, and pregnancy as well as with pathological stress (eg, myocardial infarction, pressure overload) are causative in cardiac remodeling. Metabolism-mediated changes in gene expression, metabolite signaling, and the channeling of glucose-derived carbon toward anabolic pathways seem critical for physiological growth of the heart, and metabolic inefficiency and loss of coordinated anabolic activity are emerging as proximal causes of pathological remodeling. This review integrates knowledge of different forms of cardiac remodeling to develop general models of how relationships between catabolic and anabolic glucose metabolism may fortify cardiac health or promote (mal)adaptive myocardial remodeling. Adoption of conceptual frameworks based in relational biology may enable further understanding of how metabolism regulates cardiac structure and function.
Collapse
Affiliation(s)
- Andrew A Gibb
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (A.A.G.)
| | - Bradford G Hill
- the Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville School of Medicine, KY (B.G.H.).
| |
Collapse
|
105
|
Obesity, Echocardiographic Changes and Framingham Risk Score in the Spectrum of Gout: A Cross-Sectional Study. Arch Rheumatol 2019; 34:176-185. [PMID: 31497764 DOI: 10.5606/archrheumatol.2019.7062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 08/02/2018] [Indexed: 11/21/2022] Open
Abstract
Objectives This study aims to establish cardiovascular risk in obese and non-obese patients in stages of gout by using Framingham risk score (FRS) and transthoracic echocardiography. Patients and methods This single-center cross-sectional study encompassed 201 patients (160 males, 41 females; mean age 56.9±13 years; range 20 to 89 years) including 52 asymptomatic hyperuricemia, 86 gouty arthritis without tophi, and 63 gouty tophi patients. Body Mass Index (BMI) and FRS were calculated. Left atrium (LA), interventricular septum, posterior wall (PW) of the left ventricle, fractional shortening (FS), mitral annular systolic velocity (S'), mitral annular early diastolic velocity (E') and transmitral to mitral annular early diastolic velocity ratio (E/E') were measured. Data were analyzed by Kolmogorov-Smirnov test, Shapiro-Wilk test, t-test, Mann-Whitney U test, analysis of variance test and multiple linear regression models. Results There was no significant difference in FRS, FS, S', E' and E/E' between obese and non-obese patients with asymptomatic hyperuricemia, gouty arthritis without tophi or gouty tophi. Obese patients in the three disease gradations had larger LA (p=0.007, p=0.004, p=0.039) and thicker PW (p=0.002, p=0.037, p=0.007). Increased BMI independently predicted the thickening of the PW in asymptomatic hyperuricemia (R2=0.319), gouty arthritis without tophi (R2=0.093) and gouty tophi (R2=0.068). Conclusion Despite the lack of difference in FRS and functional systolic and diastolic parameters between obese and non-obese patients in the spectrum of gout, morphological heart changes were more pronounced in obese patients. In gouty tophi, it is possible that higher urate load together with chronic inflammation contribute for the alterations, as obesity worsens them.
Collapse
|
106
|
Bonezzi F, Piccoli M, Dei Cas M, Paroni R, Mingione A, Monasky MM, Caretti A, Riganti C, Ghidoni R, Pappone C, Anastasia L, Signorelli P. Sphingolipid Synthesis Inhibition by Myriocin Administration Enhances Lipid Consumption and Ameliorates Lipid Response to Myocardial Ischemia Reperfusion Injury. Front Physiol 2019; 10:986. [PMID: 31447688 PMCID: PMC6696899 DOI: 10.3389/fphys.2019.00986] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/15/2019] [Indexed: 12/19/2022] Open
Abstract
Myocardial infarct requires prompt thrombolytic therapy or primary percutaneous coronary intervention to limit the extent of necrosis, but reperfusion creates additional damage. Along with reperfusion, a maladaptive remodeling phase might occur and it is often associated with inflammation, oxidative stress, as well as a reduced ability to recover metabolism homeostasis. Infarcted individuals can exhibit reduced lipid turnover and their accumulation in cardiomyocytes, which is linked to a deregulation of peroxisome proliferator activated receptors (PPARs), controlling fatty acids metabolism, energy production, and the anti-inflammatory response. We previously demonstrated that Myriocin can be effectively used as post-conditioning therapeutic to limit ischemia/reperfusion-induced inflammation, oxidative stress, and infarct size, in a murine model. In this follow-up study, we demonstrate that Myriocin has a critical regulatory role in cardiac remodeling and energy production, by up-regulating the transcriptional factor EB, PPARs nuclear receptors and genes involved in fatty acids metabolism, such as VLDL receptor, Fatp1, CD36, Fabp3, Cpts, and mitochondrial FA dehydrogenases. The overall effects are represented by an increased β–oxidation, together with an improved electron transport chain and energy production. The potent immunomodulatory and metabolism regulatory effects of Myriocin elicit the molecule as a promising pharmacological tool for post-conditioning therapy of myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Fabiola Bonezzi
- Stem Cells for Tissue Engineering Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Piccoli
- Stem Cells for Tissue Engineering Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Michele Dei Cas
- Clinical Biochemistry and Mass Spectrometry Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| | - Rita Paroni
- Clinical Biochemistry and Mass Spectrometry Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| | - Alessandra Mingione
- Biochemistry and Molecular Biology Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| | | | - Anna Caretti
- Biochemistry and Molecular Biology Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| | - Chiara Riganti
- Cell Biochemistry Laboratory, Oncology Department, and Interdepartmental Research Center for Molecular Biotechnology, University of Turin, Turin, Italy
| | - Riccardo Ghidoni
- Biochemistry and Molecular Biology Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| | - Carlo Pappone
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Luigi Anastasia
- Stem Cells for Tissue Engineering Laboratory, IRCCS Policlinico San Donato, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Paola Signorelli
- Biochemistry and Molecular Biology Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| |
Collapse
|
107
|
Cardiac Insulin Resistance in Heart Failure: The Role of Mitochondrial Dynamics. Int J Mol Sci 2019; 20:ijms20143552. [PMID: 31330848 PMCID: PMC6678249 DOI: 10.3390/ijms20143552] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/12/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022] Open
Abstract
Heart failure (HF) frequently coexists with conditions associated with glucose insufficiency, such as insulin resistance and type 2 diabetes mellitus (T2DM), and patients with T2DM have a significantly high incidence of HF. These two closely related diseases cannot be separated on the basis of their treatment. Some antidiabetic drugs failed to improve cardiac outcomes in T2DM patients, despite lowering glucose levels sufficiently. This may be, at least in part, due to a lack of understanding of cardiac insulin resistance. Basic investigations have revealed the significant contribution of cardiac insulin resistance to the pathogenesis and progression of HF; however, there is no clinical evidence of the definition or treatment of cardiac insulin resistance. Mitochondrial dynamics play an important role in cardiac insulin resistance and HF because they maintain cellular homeostasis through energy production, cell survival, and cell proliferation. The innovation of diagnostic tools and/or treatment targeting mitochondrial dynamics is assumed to improve not only the insulin sensitivity of the myocardium and cardiac metabolism, but also the cardiac contraction function. In this review, we summarized the current knowledge on the correlation between cardiac insulin resistance and progression of HF, and discussed the role of mitochondrial dynamics on the pathogenesis of cardiac insulin resistance and HF. We further discuss the possibility of mitochondria-targeted intervention to improve cardiac metabolism and HF.
Collapse
|
108
|
Wu KM, Hsu YM, Ying MC, Tsai FJ, Tsai CH, Chung JG, Yang JS, Tang CH, Cheng LY, Su PH, Viswanadha VP, Kuo WW, Huang CY. High-density lipoprotein ameliorates palmitic acid-induced lipotoxicity and oxidative dysfunction in H9c2 cardiomyoblast cells via ROS suppression. Nutr Metab (Lond) 2019; 16:36. [PMID: 31149020 PMCID: PMC6537189 DOI: 10.1186/s12986-019-0356-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/18/2019] [Indexed: 01/22/2023] Open
Abstract
Background High levels circulating saturated fatty acids are associated with diabetes, obesity and hyperlipidemia. In heart, the accumulation of saturated fatty acids has been determined to play a role in the development of heart failure and diabetic cardiomyopathy. High-density lipoprotein (HDL) has been reported to possess key atheroprotective biological properties, including cellular cholesterol efflux capacity, anti-oxidative and anti-inflammatory activities. However, the underlying mechanisms are still largely unknown. Therefore, the aim of the present study is to test whether HDL could protect palmitic acid (PA)-induced cardiomyocyte injury and explore the possible mechanisms. Results H9c2 cells were pretreated with HDL (50–100 μg/ml) for 2 h followed by PA (0.5 mM) for indicated time period. Our results showed that HDL inhibited PA-induced cell death in a dose-dependent manner. Moreover, HDL rescued PA-induced ROS generation and the phosphorylation of JNK which in turn activated NF-κB-mediated inflammatory proteins expressions. We also found that PA impaired the balance of BCL2 family proteins, destabilized mitochondrial membrane potential, and triggered subsequent cytochrome c release into the cytosol and activation of caspase 3. These detrimental effects were ameliorated by HDL treatment. Conclusion PA-induced ROS accumulation and results in cardiomyocyte apoptosis and inflammation. However, HDL attenuated PA-induced lipotoxicity and oxidative dysfunction via ROS suppression. These results may provide insight into a possible molecular mechanism underlying HDL suppression of the free fatty acid-induced cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Kuen-Ming Wu
- 1Department of chest medicine, Jen-Ai Hospital, Taichung, Taiwan
| | - Yuan-Man Hsu
- 2Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Mei-Chin Ying
- 3Department of Food Nutrition and Health Biotechnology, Asia University, Taichung City, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung City, Taiwan
| | - Fuu-Jen Tsai
- 5School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402 Taiwan.,6China Medical University Children's Hospital, China Medical University, Taichung, Taiwan
| | - Chang-Hai Tsai
- 6China Medical University Children's Hospital, China Medical University, Taichung, Taiwan.,7Department of Healthcare Administration, Asia University, Taichung, Taiwan
| | - Jing-Gung Chung
- 2Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- 9Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,10Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Li-Yi Cheng
- 11Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Po-Hua Su
- 12Department of Radiology, Jen-Ai Hospital, Taichung, Taiwan
| | | | - Wei-Wen Kuo
- 2Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.,14Department of Biotechnology, Asia University, Taichung, Taiwan.,15College of Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
109
|
Jensen CF, Bartels ED, Braunstein TH, Nielsen LB, Holstein‐Rathlou N, Axelsen LN, Nielsen MS. Acute intramyocardial lipid accumulation in rats does not slow cardiac conduction per se. Physiol Rep 2019; 7:e14049. [PMID: 30968589 PMCID: PMC6456446 DOI: 10.14814/phy2.14049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 01/14/2023] Open
Abstract
Diabetic patients suffer from both cardiac lipid accumulation and an increased risk of arrhythmias and sudden cardiac death. This correlation suggests a link between diabetes induced cardiac steatosis and electrical abnormalities, however, the underlying mechanism remains unknown. We previously showed that cardiac conduction velocity slows in Zucker diabetic fatty rats and in fructose-fat fed rats, models that both exhibit prominent cardiac steatosis. The aim of this study was to investigate whether acute cardiac lipid accumulation reduces conduction velocity per se. Cardiac lipid accumulation was induced acutely by perfusing isolated rat hearts with palmitate-glucose buffer, or subacutely by fasting rats overnight. Subsequently, longitudinal cardiac conduction velocity was measured in right ventricular tissue strips, and intramyocardial triglyceride and lipid droplet content was determined by thin layer chromatography and BODIPY staining, respectively. Perfusion with palmitate-glucose buffer significantly increased intramyocardial triglyceride levels compared to perfusion with glucose (2.16 ± 0.17 (n = 10) vs. 0.92 ± 0.33 nmol/mg WW (n = 9), P < 0.01), but the number of lipid droplets was very low in both groups. Fasting of rats, however, resulted in both significantly elevated intramyocardial triglyceride levels compared to fed rats (3.27 ± 0.43 (n = 10) vs. 1.45 ± 0.24 nmol/mg WW (n = 10)), as well as a larger volume of lipid droplets (0.60 ± 0.13 (n = 10) vs. 0.21 ± 0.06% (n = 10), P < 0.05). There was no significant difference in longitudinal conduction velocity between palmitate-glucose perfused and control hearts (0.77 ± 0.025 (n = 10) vs. 0.75 m/sec ± 0.029 (n = 9)), or between fed and fasted rats (0.75 ± 0.042 m/sec (n = 10) vs. 0.79 ± 0.047 (n = 10)). In conclusion, intramyocardial lipid accumulation does not slow cardiac longitudinal conduction velocity per se. This is true for both increased intramyocardial triglyceride content, induced by palmitate-glucose perfusion, and increased intramyocardial triglyceride and lipid droplet content, generated by fasting.
Collapse
Affiliation(s)
- Christa F. Jensen
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Emil D. Bartels
- Department of Clinical BiochemistryCopenhagen University Hospital RigshospitaletCopenhagenDenmark
| | - Thomas H. Braunstein
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Lars B. Nielsen
- Department of Clinical BiochemistryCopenhagen University Hospital RigshospitaletCopenhagenDenmark
| | | | - Lene N. Axelsen
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Morten Schak Nielsen
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
110
|
Goldenberg JR, Carley AN, Ji R, Zhang X, Fasano M, Schulze PC, Lewandowski ED. Preservation of Acyl Coenzyme A Attenuates Pathological and Metabolic Cardiac Remodeling Through Selective Lipid Trafficking. Circulation 2019; 139:2765-2777. [PMID: 30909726 DOI: 10.1161/circulationaha.119.039610] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Metabolic remodeling in heart failure contributes to dysfunctional lipid trafficking and lipotoxicity. Acyl coenzyme A synthetase-1 (ACSL1) facilitates long-chain fatty acid (LCFA) uptake and activation with coenzyme A (CoA), mediating the fate of LCFA. The authors tested whether cardiac ACSL1 overexpression aids LCFA oxidation and reduces lipotoxicity under pathological stress of transverse aortic constriction (TAC). METHODS Mice with cardiac restricted ACSL1 overexpression (MHC-ACSL1) underwent TAC or sham surgery followed by serial in vivo echocardiography for 14 weeks. At the decompensated stage of hypertrophy, isolated hearts were perfused with 13C LCFA during dynamic-mode 13C nuclear magnetic resonance followed by in vitro nuclear magnetic resonance and mass spectrometry analysis to assess intramyocardial lipid trafficking. In parallel, acyl CoA was measured in tissue obtained from heart failure patients pre- and postleft ventricular device implantation plus matched controls. RESULTS TAC-induced cardiac hypertrophy and dysfunction was mitigated in MHC-ACSL1 hearts compared with nontransgenic hearts. At 14 weeks, TAC increased heart weight to tibia length by 46% in nontransgenic mice, but only 26% in MHC-ACSL1 mice, whereas ACSL1 mice retained greater ejection fraction (ACSL1 TAC: 65.8±7.5%; nontransgenic TAC: 45.9±7.3) and improvement in diastolic E/E'. Functional improvements were mediated by ACSL1 changes to cardiac LCFA trafficking. ACSL1 accelerated LCFA uptake, preventing C16 acyl CoA loss post-TAC. Long-chain acyl CoA was similarly reduced in human failing myocardium and restored to control levels by mechanical unloading. ACSL1 trafficked LCFA into ceramides without normalizing the reduced triglyceride storage in TAC. ACSL1 prevented de novo synthesis of cardiotoxic C16- and C24-, and C24:1 ceramides and increased potentially cardioprotective C20- and C22-ceramides post-TAC. ACLS1 overexpression activated AMP activated protein kinase at baseline, but during TAC, prevented the reduced LCFA oxidation in hypertrophic hearts and normalized energy state (phosphocreatine:ATP) and consequently, AMP activated protein kinase activation. CONCLUSIONS This is the first demonstration of reduced acyl CoA in failing hearts of humans and mice, and suggests possible mechanisms for maintaining mitochondrial oxidative energy metabolism by restoring long-chain acyl CoA through ASCL1 activation and mechanical unloading. By mitigating cardiac lipotoxicity, via redirected LCFA trafficking to ceramides, and restoring acyl CoA, ACSL1 delayed progressive cardiac remodeling and failure.
Collapse
Affiliation(s)
- Joseph R Goldenberg
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago (J.R.G., E.D.L.)
| | - Andrew N Carley
- Department of Internal Medicine, College of Medicine, The Ohio State University (A.N.C., M.F., E.D.L.), Columbus.,Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center (A.N.C., M.F., E.D.L.), Columbus
| | - Ruiping Ji
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York (R.J., X.Z., P.C.S.)
| | - Xiaokan Zhang
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York (R.J., X.Z., P.C.S.)
| | - Matt Fasano
- Department of Internal Medicine, College of Medicine, The Ohio State University (A.N.C., M.F., E.D.L.), Columbus.,Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center (A.N.C., M.F., E.D.L.), Columbus
| | - P Christian Schulze
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York (R.J., X.Z., P.C.S.).,Department of Medicine I, Division of Cardiology, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (P.C.S.)
| | - E Douglas Lewandowski
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago (J.R.G., E.D.L.).,Department of Internal Medicine, College of Medicine, The Ohio State University (A.N.C., M.F., E.D.L.), Columbus.,Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center (A.N.C., M.F., E.D.L.), Columbus
| |
Collapse
|
111
|
Bartels ED, Guo S, Kousholt BS, Larsen JR, Hasenkam JM, Burnett J, Nielsen LB, Ashina M, Goetze JP. High doses of ANP and BNP exacerbate lipolysis in humans and the lipolytic effect of BNP is associated with cardiac triglyceride content in pigs. Peptides 2019; 112:43-47. [PMID: 30508635 DOI: 10.1016/j.peptides.2018.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022]
Abstract
Drugs facilitating the cardioprotective effects of natriuretic peptides are introduced in heart failure treatment. ANP and BNP also stimulate lipolysis and increase circulating concentrations of free fatty acids (FFAs); an aspect, however, thought to be confined to primates. We examined the lipolytic effect of natriuretic peptide infusion in healthy young men and evaluated the effect in a porcine model of myocardial ischemia and reperfusion. Six young healthy normotensive men underwent infusion with ANP, BNP, or CNP for 20 min. Blood samples were collected before, during, and after infusion for measurement of FFAs. In a porcine model of myocardial ischemia and reperfusion, animals were infused for 3 h with either BNP (n = 7) or saline (n = 5). Blood samples were collected throughout the infusion period, and cardiac tissue was obtained after infusion for lipid analysis. In humans, ANP infusion dose-dependently increased the FFA concentration in plasma 2.5-10-fold (baseline vs. 0.05 μg/kg/min P < 0.002) and with BNP 1.6-3.5-fold (P = 0.001, baseline vs. 0.02 μg/kg/min) 30 min after initiation of infusion. Infusion of CNP did not affect plasma FFA. In pigs, BNP infusion induced a 3.5-fold increase in plasma FFA (P < 0.0001), which remained elevated throughout the infusion period. Triglyceride content in porcine right cardiac ventricle tissue increased ∼5.5 fold in animals infused with BNP (P = 0.02). Natriuretic peptide infusion has similar lipolytic activity in human and pig. Our data suggest that short-term infusion increases the cardiac lipid content, and that the pig is a suitable model for studies of long-term effects mediated by natriuretic peptides.
Collapse
Affiliation(s)
- Emil D Bartels
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Song Guo
- Department of Neurology and Danish Headache Center, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte S Kousholt
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Jens R Larsen
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - J Michael Hasenkam
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - John Burnett
- Department of Cardiorenal physiology (Mayo Clinic, Rochester, MN, USA
| | - Lars B Nielsen
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Copenhagen University, Denmark; Aarhus University, Denmark
| | - Messoud Ashina
- Department of Neurology and Danish Headache Center, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen University, Denmark
| | - Jens P Goetze
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Cardiorenal physiology (Mayo Clinic, Rochester, MN, USA; Copenhagen University, Denmark
| |
Collapse
|
112
|
Obesity cardiomyopathy: the role of obstructive sleep apnea and obesity hypoventilation syndrome. Ir J Med Sci 2019; 188:783-790. [DOI: 10.1007/s11845-018-01959-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 12/19/2018] [Indexed: 01/03/2023]
|
113
|
Shang C, Sun W, Wang C, Wang X, Zhu H, Wang L, Yang H, Wang X, Gong F, Pan H. Comparative Proteomic Analysis of Visceral Adipose Tissue in Morbidly Obese and Normal Weight Chinese Women. Int J Endocrinol 2019; 2019:2302753. [PMID: 31929791 PMCID: PMC6935805 DOI: 10.1155/2019/2302753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/26/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Visceral adipose tissue (VAT) plays a central role in the balance of energy metabolism. The objective of this study was to investigate the differentially expressed proteins in VAT between morbidly obese (BMI >35 kg/m2) and normal weight Chinese women. METHOD Nine morbidly obese women and 8 normal weight women as controls were enrolled. Abdominal VAT was excised and analyzed by label-free one-dimensional liquid chromatography tandem mass spectrometry (1D-LC-MS/MS). Differentially expressed VAT proteins were further analyzed with Gene Ontology (GO) analysis and Ingenuity Pathway Analysis (IPA). Masson's trichrome staining and CD68 immunohistochemical staining of VAT were conducted in all subjects. RESULT A total of 124 differentially expressed proteins were found with a ≥2-fold difference. Forty-one proteins were upregulated, and 83 proteins were downregulated in obese individuals. These altered VAT proteins were involved in the attenuation of the liver X receptor/retinoid X receptor (LXR/RXR) signaling pathway and the activation of the acute-phase response process. Three proteins (ACSL1, HADH, and UCHL1) were validated by western blotting using the same set of VAT samples from 6 morbidly obese and 7 normal weight patients, and the results indicated that the magnitude and direction of the protein changes were in accordance with the proteomic analysis. Masson's trichrome staining and CD68 immunohistochemical staining demonstrated that there was much more collagen fiber deposition and CD68-positive macrophages in the VAT of morbidly obese patients, suggesting extensive fiber deposition and macrophage infiltration. CONCLUSION A number of differentially expressed proteins were identified in VAT between morbidly obese and normal weight Chinese females. These differential proteins could be potential candidates in addressing the role of VAT in the development of obesity.
Collapse
Affiliation(s)
- Chen Shang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100730, China
| | - Wei Sun
- Core Facility of Instrument, Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Chunlin Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100730, China
| | - Xiangqing Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100730, China
| | - Huijuan Zhu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100730, China
| | - Linjie Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100730, China
| | - Hongbo Yang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100730, China
| | - Xue Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100730, China
| | - Fengying Gong
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100730, China
| | - Hui Pan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
114
|
Mahmod M, Pal N, Rayner J, Holloway C, Raman B, Dass S, Levelt E, Ariga R, Ferreira V, Banerjee R, Schneider JE, Rodgers C, Francis JM, Karamitsos TD, Frenneaux M, Ashrafian H, Neubauer S, Rider O. The interplay between metabolic alterations, diastolic strain rate and exercise capacity in mild heart failure with preserved ejection fraction: a cardiovascular magnetic resonance study. J Cardiovasc Magn Reson 2018; 20:88. [PMID: 30580760 PMCID: PMC6304764 DOI: 10.1186/s12968-018-0511-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 11/27/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Heart failure (HF) is characterized by altered myocardial substrate metabolism which can lead to myocardial triglyceride accumulation (steatosis) and lipotoxicity. However its role in mild HF with preserved ejection fraction (HFpEF) is uncertain. We measured myocardial triglyceride content (MTG) in HFpEF and assessed its relationships with diastolic function and exercise capacity. METHODS Twenty seven HFpEF (clinical features of HF, left ventricular EF >50%, evidence of mild diastolic dysfunction and evidence of exercise limitation as assessed by cardiopulmonary exercise test) and 14 controls underwent 1H-cardiovascular magnetic resonance spectroscopy (1H-CMRS) to measure MTG (lipid/water, %), 31P-CMRS to measure myocardial energetics (phosphocreatine-to-adenosine triphosphate - PCr/ATP) and feature-tracking cardiovascular magnetic resonance (CMR) imaging for diastolic strain rate. RESULTS When compared to controls, HFpEF had 2.3 fold higher in MTG (1.45 ± 0.25% vs. 0.64 ± 0.16%, p = 0.009) and reduced PCr/ATP (1.60 ± 0.09 vs. 2.00 ± 0.10, p = 0.005). HFpEF had significantly reduced diastolic strain rate and maximal oxygen consumption (VO2 max), which both correlated significantly with elevated MTG and reduced PCr/ATP. On multivariate analyses, MTG was independently associated with diastolic strain rate while diastolic strain rate was independently associated with VO2 max. CONCLUSIONS Myocardial steatosis is pronounced in mild HFpEF, and is independently associated with impaired diastolic strain rate which is itself related to exercise capacity. Steatosis may adversely affect exercise capacity by indirect effect occurring via impairment in diastolic function. As such, myocardial triglyceride may become a potential therapeutic target to treat the increasing number of patients with HFpEF.
Collapse
Affiliation(s)
- Masliza Mahmod
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
- National University of Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Nikhil Pal
- Divisions of Experimental Therapeutics and Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Jennifer Rayner
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Cameron Holloway
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Betty Raman
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Sairia Dass
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Eylem Levelt
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Rina Ariga
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Vanessa Ferreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Rajarshi Banerjee
- 1st Department of Cardiology, Aristotle University, Thessaloniki, Greece
| | - Jurgen E. Schneider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Christopher Rodgers
- Department of Medicine, John Radcliffe Hospital, Oxford, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Jane M. Francis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Theodoros D. Karamitsos
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
- 1st Department of Cardiology, Aristotle University, Thessaloniki, Greece
| | - Michael Frenneaux
- Norwich Medical School, Bob Champion Research and Education Building, James Watson Road, University of East Anglia Norwich Research Park, Norwich, NR4 7UQ UK
| | - Houman Ashrafian
- Divisions of Experimental Therapeutics and Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Oliver Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| |
Collapse
|
115
|
Fang CY, Chen MC, Chang TH, Wu CC, Chang JP, Huang HD, Ho WC, Wang YZ, Pan KL, Lin YS, Huang YK, Chen CJ, Lee WC. Idi1 and Hmgcs2 Are Affected by Stretch in HL-1 Atrial Myocytes. Int J Mol Sci 2018; 19:ijms19124094. [PMID: 30567295 PMCID: PMC6321625 DOI: 10.3390/ijms19124094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 01/27/2023] Open
Abstract
Background: Lipid expression is increased in the atrial myocytes of mitral regurgitation (MR) patients. This study aimed to investigate key regulatory genes and mechanisms of atrial lipotoxic myopathy in MR. Methods: The HL-1 atrial myocytes were subjected to uniaxial cyclic stretching for eight hours. Fatty acid metabolism, lipoprotein signaling, and cholesterol metabolism were analyzed by PCR assay (168 genes). Results: The stretched myocytes had significantly larger cell size and higher lipid expression than non-stretched myocytes (all p < 0.001). Fatty acid metabolism, lipoprotein signaling, and cholesterol metabolism in the myocytes were analyzed by PCR assay (168 genes). In comparison with their counterparts in non-stretched myocytes, seven genes in stretched monocytes (Idi1, Olr1, Nr1h4, Fabp2, Prkag3, Slc27a5, Fabp6) revealed differential upregulation with an altered fold change >1.5. Nine genes in stretched monocytes (Apoa4, Hmgcs2, Apol8, Srebf1, Acsm4, Fabp1, Acox2, Acsl6, Gk) revealed differential downregulation with an altered fold change <0.67. Canonical pathway analysis, using Ingenuity Pathway Analysis software, revealed that the only genes in the “superpathway of cholesterol biosynthesis” were Idi1 (upregulated) and Hmgcs2 (downregulated). The fraction of stretched myocytes expressing Nile red was significantly decreased by RNA interference of Idi1 (p < 0.05) and was significantly decreased by plasmid transfection of Hmgcs2 (p = 0.004). Conclusions: The Idi1 and Hmgcs2 genes have regulatory roles in atrial lipotoxic myopathy associated with atrial enlargement.
Collapse
Affiliation(s)
- Chih-Yuan Fang
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Mien-Cheng Chen
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei 110, Taiwan.
| | - Chia-Chen Wu
- Division of Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Jen-Ping Chang
- Division of Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Hsien-Da Huang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu 300, Taiwan.
| | - Wan-Chun Ho
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Yi-Zhen Wang
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Kuo-Li Pan
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Yu-Sheng Lin
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Yao-Kuang Huang
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Chien-Jen Chen
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Wei-Chieh Lee
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| |
Collapse
|
116
|
Yamamoto T, Endo J, Kataoka M, Matsuhashi T, Katsumata Y, Shirakawa K, Yoshida N, Isobe S, Moriyama H, Goto S, Yamashita K, Nakanishi H, Shimanaka Y, Kono N, Shinmura K, Arai H, Fukuda K, Sano M. Decrease in membrane phospholipids unsaturation correlates with myocardial diastolic dysfunction. PLoS One 2018; 13:e0208396. [PMID: 30533011 PMCID: PMC6289418 DOI: 10.1371/journal.pone.0208396] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/17/2018] [Indexed: 11/19/2022] Open
Abstract
Increase in saturated fatty acid (SFA) content in membrane phospholipids dramatically affects membrane properties and cellular functioning. We sought to determine whether exogenous SFA from the diet directly affects the degree of membrane phospholipid unsaturation in adult hearts and if these changes correlate with contractile dysfunction. Although both SFA-rich high fat diets (HFDs) and monounsaturated FA (MUFA)-rich HFDs cause the same degree of activation of myocardial FA uptake, triglyceride turnover, and mitochondrial FA oxidation and accumulation of toxic lipid intermediates, the former induced more severe diastolic dysfunction than the latter, which was accompanied with a decrease in membrane phospholipid unsaturation, induction of unfolded protein response (UPR), and a decrease in the expression of Sirt1 and stearoyl-CoA desaturase-1 (SCD1), catalyzing the conversion of SFA to MUFA. When the SFA supply in the heart overwhelms the cellular capacity to use it for energy, excess exogenous SFA channels to membrane phospholipids, leading to UPR induction, and development of diastolic dysfunction.
Collapse
Affiliation(s)
- Tsunehisa Yamamoto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Jin Endo
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
- Japan Science and Technology Agency, Tokyo, Japan
| | - Masaharu Kataoka
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | | | | | - Kohsuke Shirakawa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Naohiro Yoshida
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
- Department of Endocrinology and Hypertension, Tokyo Women’s Medical University, Tokyo, Japan
| | - Sarasa Isobe
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Hidenori Moriyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shinichi Goto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Kaoru Yamashita
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
- Department of Endocrinology and Hypertension, Tokyo Women’s Medical University, Tokyo, Japan
| | | | - Yuta Shimanaka
- Graduate School of Pharmaceutical Sciences, Tokyo University, Tokyo, Japan
| | - Nozomu Kono
- Graduate School of Pharmaceutical Sciences, Tokyo University, Tokyo, Japan
| | - Ken Shinmura
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
- Department of General Medicine, Hyogo College of Medicine, Hyogo, Japan
| | - Hiroyuki Arai
- Graduate School of Pharmaceutical Sciences, Tokyo University, Tokyo, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
- Japan Science and Technology Agency, Tokyo, Japan
- * E-mail:
| |
Collapse
|
117
|
Bildirici I, Schaiff WT, Chen B, Morizane M, Oh SY, O’Brien M, Sonnenberg-Hirche C, Chu T, Barak Y, Nelson DM, Sadovsky Y. PLIN2 Is Essential for Trophoblastic Lipid Droplet Accumulation and Cell Survival During Hypoxia. Endocrinology 2018; 159:3937-3949. [PMID: 30351430 PMCID: PMC6240902 DOI: 10.1210/en.2018-00752] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Trophoblast hypoxia and injury, key components of placental dysfunction, are associated with fetal growth restriction and other complications of pregnancy. Accumulation of lipid droplets has been found in hypoxic nonplacental cells. Unique to pregnancy, lipid accumulation in the placenta might perturb lipid transport to the fetus. We tested the hypothesis that hypoxia leads to accumulation of lipid droplets in human trophoblasts and that trophoblastic PLIN proteins play a key role in this process. We found that hypoxia promotes the accumulation of lipid droplets in primary human trophoblasts. A similar accretion of lipid droplets was found in placental villi in vivo from pregnancies complicated by fetal growth restriction. In both situations, these changes were associated with an increased level of cellular triglycerides. Exposure of trophoblasts to hypoxia led to reduced fatty acid efflux and oxidation with no change in fatty acid uptake or synthesis. We further found that hypoxia markedly stimulated PLIN2 mRNA synthesis and protein expression, which colocalized to lipid droplets. Knockdown of PLIN2, but not PLIN3, enhanced trophoblast apoptotic death, and overexpression of PLIN2 promoted cell viability. Collectively, our data indicate that hypoxia enhances trophoblastic lipid retention in the form of lipid droplets and that PLIN2 plays a key role in this process and in trophoblast defense against apoptotic death. These findings also imply that this protective mechanism may lead to diminished trafficking of lipids to the developing fetus.
Collapse
Affiliation(s)
- Ibrahim Bildirici
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - W Timothy Schaiff
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Baosheng Chen
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Mayumi Morizane
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Soo-Young Oh
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew O’Brien
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Tianjiao Chu
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yaacov Barak
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - D Michael Nelson
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
- Correspondence: Yoel Sadovsky, MD, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, Pennsylvania 15213. E-mail:
| |
Collapse
|
118
|
Shete V, Liu N, Jia Y, Viswakarma N, Reddy JK, Thimmapaya B. Mouse Cardiac Pde1C Is a Direct Transcriptional Target of Pparα. Int J Mol Sci 2018; 19:ijms19123704. [PMID: 30469494 PMCID: PMC6321386 DOI: 10.3390/ijms19123704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/29/2022] Open
Abstract
Phosphodiesterase 1C (PDE1C) is expressed in mammalian heart and regulates cardiac functions by controlling levels of second messenger cyclic AMP and cyclic GMP (cAMP and cGMP, respectively). However, molecular mechanisms of cardiac Pde1c regulation are currently unknown. In this study, we demonstrate that treatment of wild type mice and H9c2 myoblasts with Wy-14,643, a potent ligand of nuclear receptor peroxisome-proliferator activated receptor alpha (PPARα), leads to elevated cardiac Pde1C mRNA and cardiac PDE1C protein, which correlate with reduced levels of cAMP. Furthermore, using mice lacking either Pparα or cardiomyocyte-specific Med1, the major subunit of Mediator complex, we show that Wy-14,643-mediated Pde1C induction fails to occur in the absence of Pparα and Med1 in the heart. Finally, using chromatin immunoprecipitation assays we demonstrate that PPARα binds to the upstream Pde1C promoter sequence on two sites, one of which is a palindrome sequence (agcTAGGttatcttaacctagc) that shows a robust binding. Based on these observations, we conclude that cardiac Pde1C is a direct transcriptional target of PPARα and that Med1 may be required for the PPARα mediated transcriptional activation of cardiac Pde1C.
Collapse
Affiliation(s)
- Varsha Shete
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Ning Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Yuzhi Jia
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Navin Viswakarma
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Janardan K Reddy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Bayar Thimmapaya
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
119
|
Riehle C, Bauersachs J. Of mice and men: models and mechanisms of diabetic cardiomyopathy. Basic Res Cardiol 2018; 114:2. [PMID: 30443826 PMCID: PMC6244639 DOI: 10.1007/s00395-018-0711-0] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/09/2018] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus increases the risk of heart failure independent of co-existing hypertension and coronary artery disease. Although several molecular mechanisms for the development of diabetic cardiomyopathy have been identified, they are incompletely understood. The pathomechanisms are multifactorial and as a consequence, no causative treatment exists at this time to modulate or reverse the molecular changes contributing to accelerated cardiac dysfunction in diabetic patients. Numerous animal models have been generated, which serve as powerful tools to study the impact of type 1 and type 2 diabetes on the heart. Despite specific limitations of the models generated, they mimic various perturbations observed in the diabetic myocardium and continue to provide important mechanistic insight into the pathogenesis underlying diabetic cardiomyopathy. This article reviews recent studies in both diabetic patients and in these animal models, and discusses novel hypotheses to delineate the increased incidence of heart failure in diabetic patients.
Collapse
Affiliation(s)
- Christian Riehle
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany.
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
| |
Collapse
|
120
|
Sletten AC, Peterson LR, Schaffer JE. Manifestations and mechanisms of myocardial lipotoxicity in obesity. J Intern Med 2018; 284:478-491. [PMID: 29331057 PMCID: PMC6045461 DOI: 10.1111/joim.12728] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Environmental and socioeconomic changes over the past thirty years have contributed to a dramatic rise in the worldwide prevalence of obesity. Heart disease is amongst the most serious health risks of obesity, with increases in both atherosclerotic coronary heart disease and heart failure among obese individuals. In this review, we focus on primary myocardial alterations in obesity that include hypertrophic remodelling and diastolic dysfunction. Obesity-associated perturbations in myocardial and systemic lipid metabolism are important contributors to cardiovascular complications of obesity. Accumulation of excess lipid in nonadipose cells of the cardiovascular system can cause cell dysfunction and cell death, a process known as lipotoxicity. Lipotoxicity has been modelled in mice using high-fat diet feeding, inbred lines with mutations in leptin receptor signalling, and in genetically engineered mice with enhanced myocardial fatty acid uptake, altered lipid droplet homoeostasis or decreased cardiac fatty acid oxidation. These studies, along with findings in cell culture model systems, indicate that the molecular pathophysiology of lipid overload involves endoplasmic reticulum stress, alterations in autophagy, de novo ceramide synthesis, oxidative stress, inflammation and changes in gene expression. We highlight recent advances that extend our understanding of the impact of obesity and altered lipid metabolism on cardiac function.
Collapse
Affiliation(s)
- A C Sletten
- Department of Medicine, Washington University, St Louis, MO, USA
| | - L R Peterson
- Department of Medicine, Washington University, St Louis, MO, USA
| | - J E Schaffer
- Department of Medicine, Washington University, St Louis, MO, USA
| |
Collapse
|
121
|
Wan S, Zhang L, Quan Y, Wei K. Resveratrol-loaded PLGA nanoparticles: enhanced stability, solubility and bioactivity of resveratrol for non-alcoholic fatty liver disease therapy. ROYAL SOCIETY OPEN SCIENCE 2018; 5:181457. [PMID: 30564426 PMCID: PMC6281916 DOI: 10.1098/rsos.181457] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/16/2018] [Indexed: 05/25/2023]
Abstract
Resveratrol (3, 4', 5-trihydroxy-trans-stilbene, RSV), a nutraceutical, has recently attracted lots of attention because of its outstanding pharmacological potential. The effects of RSV on non-alcoholic fatty liver disease (NAFLD) remain inconclusive, although a wealth of research has been done. The major obstacle presented was RSV's poor bioavailability due to its poor aqueous solubility, chemical instability and intestinal metabolism. In this study, nanotechnology was used to encapsulate RSV to enhance its stability, water solubility and bioactivity, which can be used to treat NAFLD by HepG2 hepatocytes-induced in vitro. RSV-loaded poly (d, l-lactide-co-glycolide acid) (PLGA) nanoparticles (RSV-PLGA-NPs) were prepared according to an oil/water (O/W) emulsion technique. The RSV-PLGA-NPs were of spherical morphology with an average size of 176.1 nm and a negative charge of -22.6 mV. These nanoparticles exhibited remarkable encapsulation efficiency (EE%) (97.25%) and drug loading (14.9%) for RSV. A sustained RSV release from RSV-PLGA-NPs could be achieved especially in acidic conditions when simulating transporting through the gastrointestinal tract. In addition, these nanoparticles were stable enough to store at 4°C for a least six months with unchanged EE%. Moreover, RSV-PLGA-NPs were more efficient in alleviating lipogenesis, promoting lipolysis and reducing hepatocellular proliferation than free RSV due to its improved stability, water solubility and bioactivity. These findings indicated that the RSV-PLGA-NPs provided superb and stable drug delivery with small particle size, high capsulation efficiency, well-controlled drug release, which greatly enhanced the stability, water solubility and bioactivity. Besides, the discovery that the inhibitory effect of RSV-PLGA-NPs on hepatocellular proliferation and lipid accumulation in steatotic HepG2 cells may provide a new way to study the mechanism of NAFLD. Therefore, RSV-PLGA-NPs have a promising potential for NAFLD therapy.
Collapse
Affiliation(s)
- Shuqian Wan
- School of Biological Science and Engineering, South China University of Technology, Guangzhou 510640, People's Republic of China
| | - Long Zhang
- Wenzhou Institute of Biomaterials and Engineering, CAS, Wenzhou, Zhejiang 325011, People's Republic of China
| | - Yunyun Quan
- Wenzhou Institute of Biomaterials and Engineering, CAS, Wenzhou, Zhejiang 325011, People's Republic of China
| | - Kun Wei
- School of Biological Science and Engineering, South China University of Technology, Guangzhou 510640, People's Republic of China
- Wenzhou Institute of Biomaterials and Engineering, CAS, Wenzhou, Zhejiang 325011, People's Republic of China
| |
Collapse
|
122
|
Lovric A, Granér M, Bjornson E, Arif M, Benfeitas R, Nyman K, Ståhlman M, Pentikäinen MO, Lundbom J, Hakkarainen A, Sirén R, Nieminen MS, Lundbom N, Lauerma K, Taskinen MR, Mardinoglu A, Boren J. Characterization of different fat depots in NAFLD using inflammation-associated proteome, lipidome and metabolome. Sci Rep 2018; 8:14200. [PMID: 30242179 PMCID: PMC6155005 DOI: 10.1038/s41598-018-31865-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 08/21/2018] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is recognized as a liver manifestation of metabolic syndrome, accompanied with excessive fat accumulation in the liver and other vital organs. Ectopic fat accumulation was previously associated with negative effects at the systemic and local level in the human body. Thus, we aimed to identify and assess the predictive capability of novel potential metabolic biomarkers for ectopic fat depots in non-diabetic men with NAFLD, using the inflammation-associated proteome, lipidome and metabolome. Myocardial and hepatic triglycerides were measured with magnetic spectroscopy while function of left ventricle, pericardial and epicardial fat, subcutaneous and visceral adipose tissue were measured with magnetic resonance imaging. Measured ectopic fat depots were profiled and predicted using a Random Forest algorithm, and by estimating the Area Under the Receiver Operating Characteristic curves. We have identified distinct metabolic signatures of fat depots in the liver (TAG50:1, glutamate, diSM18:0 and CE20:3), pericardium (N-palmitoyl-sphinganine, HGF, diSM18:0, glutamate, and TNFSF14), epicardium (sphingomyelin, CE20:3, PC38:3 and TNFSF14), and myocardium (CE20:3, LAPTGF-β1, glutamate and glucose). Our analyses highlighted non-invasive biomarkers that accurately predict ectopic fat depots, and reflect their distinct metabolic signatures in subjects with NAFLD.
Collapse
Affiliation(s)
- Alen Lovric
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Marit Granér
- Heart and Lung Center, Division of Cardiology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Elias Bjornson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.,Department of Molecular and Clinical Medicine/Wallenberg Lab, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Muhammad Arif
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Rui Benfeitas
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Kristofer Nyman
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine/Wallenberg Lab, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Markku O Pentikäinen
- Heart and Lung Center, Division of Cardiology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Jesper Lundbom
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Antti Hakkarainen
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Reijo Sirén
- Department of General Practice and Primary Health Care, Health Care Centre of City of Helsinki and University of Helsinki, Helsinki, Finland
| | - Markku S Nieminen
- Heart and Lung Center, Division of Cardiology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Nina Lundbom
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Kirsi Lauerma
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Marja-Riitta Taskinen
- Heart and Lung Center, Division of Cardiology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland.
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden. .,Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
| | - Jan Boren
- Department of Molecular and Clinical Medicine/Wallenberg Lab, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
123
|
Liu Y, Neumann D, Glatz JFC, Luiken JJFP. Molecular mechanism of lipid-induced cardiac insulin resistance and contractile dysfunction. Prostaglandins Leukot Essent Fatty Acids 2018; 136:131-141. [PMID: 27372802 DOI: 10.1016/j.plefa.2016.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/10/2016] [Indexed: 01/04/2023]
Abstract
Long-chain fatty acids are the main cardiac substrates from which ATP is generated continually to serve the high energy demand and sustain the normal function of the heart. Under healthy conditions, fatty acid β-oxidation produces 50-70% of the energy demands with the remainder largely accounted for by glucose. Chronically increased dietary lipid supply often leads to excess lipid accumulation in the heart, which is linked to a variety of maladaptive phenomena, such as insulin resistance, cardiac hypertrophy and contractile dysfunction. CD36, the predominant cardiac fatty acid transporter, has a key role in setting the heart on a road to contractile dysfunction upon the onset of chronic lipid oversupply by translocating to the cell surface and opening the cellular 'doors' for fatty acids. The sequence of events after the CD36-mediated myocellular lipid accumulation is less understood, but in general it has been accepted that the excessively imported lipids cause insulin resistance, which in turn leads to contractile dysfunction. There are several gaps of knowledge in this proposed order of events which this review aims to discuss. First, the molecular mechanisms underlying lipid-induced insulin resistance are not yet completely disclosed. Specifically, several mediators have been proposed, such as diacylglycerols, ceramides, peroxisome proliferator-activated receptors (PPAR), inflammatory kinases and reactive oxygen species (ROS), but their relative contributions to the onset of insulin resistance and their putatively synergistic actions are topics of controversy. Second, there are also pieces of evidence that lipids can induce contractile dysfunction independently of insulin resistance. Perhaps, a more integrative view is needed, in which several lipid-induced pathways operate synergistically or in parallel to induce contractile dysfunction. Unraveling of these processes is expected to be important in designing effective therapeutic strategies to protect the lipid-overloaded heart.
Collapse
Affiliation(s)
- Yilin Liu
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Dietbert Neumann
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Jan F C Glatz
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Joost J F P Luiken
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
124
|
Xu Y, Zhang Y, Ye J. IL-6: A Potential Role in Cardiac Metabolic Homeostasis. Int J Mol Sci 2018; 19:ijms19092474. [PMID: 30134607 PMCID: PMC6164544 DOI: 10.3390/ijms19092474] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022] Open
Abstract
Interleukin-6 (IL-6) is implicated in multiple biological functions including immunity, neural development, and haematopoiesis. Recently, mounting evidence indicates that IL-6 plays a key role in metabolism, especially lipid metabolic homeostasis. A working heart requires a high and constant energy input which is largely generated by fatty acid (FA) β-oxidation. Under pathological conditions, the precise balance between cardiac FA uptake and metabolism is perturbed so that excessive FA is accumulated, thereby predisposing to myocardial dysfunction (cardiac lipotoxicity). In this review, we summarize the current evidence that suggests the involvement of IL-6 in lipid metabolism. Cardiac metabolic features and consequences of myocardial lipotoxicity are also briefly analyzed. Finally, the roles of IL-6 in cardiac FA uptake (i.e., serum lipid profile and myocardial FA transporters) and FA metabolism (namely, β-oxidation, mitochondrial function, biogenesis, and FA de novo synthesis) are discussed. Overall, understanding how IL-6 transmits signals to affect lipid metabolism in the heart might allow for development of better clinical therapies for obesity-associated cardiac lipotoxicity.
Collapse
Affiliation(s)
- Yitao Xu
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London W120NN, UK.
| | - Yubin Zhang
- State Key laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China.
| | - Junmei Ye
- State Key laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China.
| |
Collapse
|
125
|
Redondo-Angulo I, Mas-Stachurska A, Sitges M, Tinahones FJ, Giralt M, Villarroya F, Planavila A. Fgf21 is required for cardiac remodeling in pregnancy. Cardiovasc Res 2018; 113:1574-1584. [PMID: 28472473 DOI: 10.1093/cvr/cvx088] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 05/03/2017] [Indexed: 11/13/2022] Open
Abstract
Aims Fibroblast growth factor-21 (Fgf21) is an endocrine factor that contributes to many physiological and pathological processes, mainly via its action as a metabolic regulator. Recent studies have shown that Fgf21 plays an important role in cardiac tissue. Pregnancy offers a physiological model of adaptive and reversible heart enlargement, but the molecular mechanisms underlying this cardiac hypertrophy are poorly understood. Therefore, the aim was to analyze the role of Fgf21 during late pregnancy, and assess the physiological relevance of Fgf21 for cardiac tissue during this process. Methods and results Female mice and rats at day 18 of gestation and pregnant women in their third trimester were used as models of late pregnancy, and our results revealed that their plasma levels of Fgf21 were significantly increased relative to non-pregnant controls. Pregnant wild-type (wt) mice exhibited a PPARα (peroxisome proliferator-activated receptor-α)-dependent enhancement of Fgf21 expression in the liver and heart. Moreover, pregnancy altered the levels of Fgf21 receptor-1 (FGFR1) and β-klotho, and activated intracellular Fgf21 signaling in the heart. Fgf21-/- mice did not develop the pregnancy-induced cardiac remodeling seen in wt mice. Furthermore, the hearts of Fgf21-/- mice exhibited reductions in their fatty acid oxidation levels, which may compromise cardiac function during pregnancy. Conclusions During pregnancy, both systemic and cardiac-produced Fgf21 act on the heart, leading to the normal physiological cardiac changes that are associated with pregnancy. Thus, Fgf21 acts as an endocrine/autocrine factor required for cardiac remodeling response to gestation.
Collapse
Affiliation(s)
- Ibon Redondo-Angulo
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Barcelona, Spain
| | - Aleksandra Mas-Stachurska
- Cardiology Department, Thorax Institute, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| | - Marta Sitges
- Cardiology Department, Thorax Institute, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| | - Francisco José Tinahones
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Barcelona, Spain.,Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Teatinos Campus, 29010 Malaga, Spain.,Investigation Unit (IBIMA), Virgen de la Victoria Hospital, 29010 Malaga, Spain
| | - Marta Giralt
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Barcelona, Spain
| | - Francesc Villarroya
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Barcelona, Spain
| | - Anna Planavila
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Barcelona, Spain
| |
Collapse
|
126
|
A Reduction in ADAM17 Expression Is Involved in the Protective Effect of the PPAR- α Activator Fenofibrate on Pressure Overload-Induced Cardiac Hypertrophy. PPAR Res 2018; 2018:7916953. [PMID: 30105051 PMCID: PMC6076894 DOI: 10.1155/2018/7916953] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/27/2018] [Accepted: 07/08/2018] [Indexed: 12/21/2022] Open
Abstract
The peroxisome proliferator-activated receptor-α (PPAR-α) agonist fenofibrate ameliorates cardiac hypertrophy; however, its mechanism of action has not been completely determined. Our previous study indicated that a disintegrin and metalloproteinase-17 (ADAM17) is required for angiotensin II-induced cardiac hypertrophy. This study aimed to determine whether ADAM17 is involved in the protective action of fenofibrate against cardiac hypertrophy. Abdominal artery constriction- (AAC-) induced hypertensive rats were used to observe the effects of fenofibrate on cardiac hypertrophy and ADAM17 expression. Primary cardiomyocytes were pretreated with fenofibrate (10 μM) for 1 hour before being stimulated with angiotensin II (100 nM) for another 24 hours. Fenofibrate reduced the ratios of left ventricular weight to body weight (LVW/BW) and heart weight to body weight (HW/BW), left ventricular anterior wall thickness (LVAW), left ventricular posterior wall thickness (LVPW), and ADAM17 mRNA and protein levels in left ventricle in AAC-induced hypertensive rats. Similarly, in vitro experiments showed that fenofibrate significantly attenuated angiotensin II-induced cardiac hypertrophy and diminished ADAM17 mRNA and protein levels in primary cardiomyocytes stimulated with angiotensin II. In summary, a reduction in ADAM17 expression is associated with the protective action of PPAR-α agonists against pressure overload-induced cardiac hypertrophy.
Collapse
|
127
|
Karwi QG, Uddin GM, Ho KL, Lopaschuk GD. Loss of Metabolic Flexibility in the Failing Heart. Front Cardiovasc Med 2018; 5:68. [PMID: 29928647 PMCID: PMC5997788 DOI: 10.3389/fcvm.2018.00068] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/18/2018] [Indexed: 12/15/2022] Open
Abstract
To maintain its high energy demand the heart is equipped with a highly complex and efficient enzymatic machinery that orchestrates ATP production using multiple energy substrates, namely fatty acids, carbohydrates (glucose and lactate), ketones and amino acids. The contribution of these individual substrates to ATP production can dramatically change, depending on such variables as substrate availability, hormonal status and energy demand. This "metabolic flexibility" is a remarkable virtue of the heart, which allows utilization of different energy substrates at different rates to maintain contractile function. In heart failure, cardiac function is reduced, which is accompanied by discernible energy metabolism perturbations and impaired metabolic flexibility. While it is generally agreed that overall mitochondrial ATP production is impaired in the failing heart, there is less consensus as to what actual switches in energy substrate preference occur. The failing heart shift toward a greater reliance on glycolysis and ketone body oxidation as a source of energy, with a decrease in the contribution of glucose oxidation to mitochondrial oxidative metabolism. The heart also becomes insulin resistant. However, there is less consensus as to what happens to fatty acid oxidation in heart failure. While it is generally believed that fatty acid oxidation decreases, a number of clinical and experimental studies suggest that fatty acid oxidation is either not changed or is increased in heart failure. Of importance, is that any metabolic shift that does occur has the potential to aggravate cardiac dysfunction and the progression of the heart failure. An increasing body of evidence shows that increasing cardiac ATP production and/or modulating cardiac energy substrate preference positively correlates with heart function and can lead to better outcomes. This includes increasing glucose and ketone oxidation and decreasing fatty acid oxidation. In this review we present the physiology of the energy metabolism pathways in the heart and the changes that occur in these pathways in heart failure. We also look at the interventions which are aimed at manipulating the myocardial metabolic pathways toward more efficient substrate utilization which will eventually improve cardiac performance.
Collapse
Affiliation(s)
| | | | | | - Gary D. Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
128
|
Guo Y, Wang Z, Qin X, Xu J, Hou Z, Yang H, Mao X, Xing W, Li X, Zhang X, Gao F. Enhancing fatty acid utilization ameliorates mitochondrial fragmentation and cardiac dysfunction via rebalancing optic atrophy 1 processing in the failing heart. Cardiovasc Res 2018; 114:979-991. [PMID: 29490017 DOI: 10.1093/cvr/cvy052] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/24/2018] [Indexed: 09/12/2024] Open
Abstract
AIMS Heart failure (HF) is characterized by reduced fatty acid (FA) utilization associated with mitochondrial dysfunction. Recent evidence has shown that enhancing FA utilization may provide cardioprotection against HF. Our aim was to investigate the effects and the underlying mechanisms of cardiac FA utilization on cardiac function in response to pressure overload. METHODS AND RESULTS Transverse aortic constriction (TAC) was used in C57 mice to establish pressure overload-induced HF. TAC mice fed on a high fat diet (HFD) exhibited increased cardiac FA utilization and improved cardiac function and survival compared with those on control diet. Such cardioprotection could also be provided by cardiac-specific overexpression of CD36. Notably, both HFD and CD36 overexpression attenuated mitochondrial fragmentation and improved mitochondrial function in the failing heart. Pressure overload decreased ATP-dependent metalloprotease (YME1L) expression and induced the proteolytic cleavage of the dynamin-like guanosine triphosphatase OPA1 as a result of suppressed FA utilization. Enhancing FA utilization upregulated YME1L expression and subsequently rebalanced OPA1 processing, resulting in restoration of mitochondrial morphology in the failing heart. In addition, cardiac-specific overexpression of YME1L exerted similar cardioprotective effects against HF to those provided by HFD or CD36 overexpression. CONCLUSIONS These findings demonstrate that enhancing FA utilization ameliorates mitochondrial fragmentation and cardiac dysfunction via rebalancing OPA1 processing in pressure overload-induced HF, suggesting a unique metabolic intervention approach to improving cardiac functions in HF.
Collapse
MESH Headings
- Animals
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- Cells, Cultured
- Diet, High-Fat
- Disease Models, Animal
- Energy Metabolism
- Fatty Acids/metabolism
- GTP Phosphohydrolases/genetics
- GTP Phosphohydrolases/metabolism
- Heart Failure/diet therapy
- Heart Failure/metabolism
- Heart Failure/pathology
- Heart Failure/physiopathology
- Male
- Metalloendopeptidases/genetics
- Metalloendopeptidases/metabolism
- Mice, Inbred C57BL
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Proteolysis
- Rats, Sprague-Dawley
- Ventricular Function, Left
Collapse
Affiliation(s)
- Yongzheng Guo
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Zhen Wang
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Xinghua Qin
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Jie Xu
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Zuoxu Hou
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Hongyan Yang
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Xuechao Mao
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Wenjuan Xing
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Xiaoliang Li
- Department of Emergency Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xing Zhang
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| | - Feng Gao
- School of Aerospace Medicine, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China and
| |
Collapse
|
129
|
Toedebusch R, Belenchia A, Pulakat L. Diabetic Cardiomyopathy: Impact of Biological Sex on Disease Development and Molecular Signatures. Front Physiol 2018; 9:453. [PMID: 29773993 PMCID: PMC5943496 DOI: 10.3389/fphys.2018.00453] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/11/2018] [Indexed: 12/14/2022] Open
Abstract
Diabetic cardiomyopathy refers to a unique set of heart-specific pathological variables induced by hyperglycemia and insulin resistance. Given that cardiovascular disease (CVD) is the leading cause of death in the world, and type 2 diabetes incidence continues to rise, understanding the complex interplay between these two morbidities and developing novel therapeutic strategies is vital. Two hallmark characteristics specific to diabetic cardiomyopathy are diastolic dysfunction and cardiac structural mal-adaptations, arising from cardiac cellular responses to the complex toxicity induced by hyperglycemia with or without hyperinsulinemia. While type 2 diabetes is more prevalent in men compared to women, cardiovascular risk is higher in diabetic women than in diabetic men, suggesting that diabetic women take a steeper path to cardiomyopathy and heart failure. Accumulating evidence from randomized clinical trials indicate that although pre-menopausal women have lower risk of CVDs, compared to age-matched men, this advantage is lost in diabetic pre-menopausal women, which suggests estrogen availability does not protect from increased cardiovascular risk. Notably, few human studies have assessed molecular and cellular mechanisms regarding similarities and differences in the progression of diabetic cardiomyopathy in men versus women. Additionally, most pre-clinical rodent studies fail to include female animals, leaving a void in available data to truly understand the impact of biological sex differences in diabetes-induced dysfunction of cardiovascular cells. Elegant reviews in the past have discussed in detail the roles of estrogen-mediated signaling in cardiovascular protection, sex differences associated with telomerase activity in the heart, and cardiac responses to exercise. In this review, we focus on the emerging cellular and molecular markers that define sex differences in diabetic cardiomyopathy based on the recent clinical and pre-clinical evidence. We also discuss miR-208a, MED13, and AT2R, which may provide new therapeutic targets with hopes to develop novel treatment paradigms to treat diabetic cardiomyopathy uniquely between men and women.
Collapse
Affiliation(s)
- Ryan Toedebusch
- Cardiovascular Medicine Division, Department of Medicine, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Anthony Belenchia
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Lakshmi Pulakat
- Cardiovascular Medicine Division, Department of Medicine, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
130
|
Xu S, Zhang X, Liu P. Lipid droplet proteins and metabolic diseases. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1968-1983. [DOI: 10.1016/j.bbadis.2017.07.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
|
131
|
|
132
|
Recovery of Cardiac Remodeling and Dysmetabolism by Pancreatic Islet Injury Improvement in Diabetic Rats after Yacon Leaf Extract Treatment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1821359. [PMID: 30057670 PMCID: PMC6051012 DOI: 10.1155/2018/1821359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/12/2018] [Indexed: 02/06/2023]
Abstract
Yacon (Smallanthus sonchifolius) is a native Andean plant rich in phenolic compounds, and its effects on dysmetabolism and cardiomyopathy in diabetic rats was evaluated. The rats (10/group) were allocated as follows: C, controls; C + Y, controls treated with Yacon leaf extract (YLE); DM, diabetic controls; and DM + Y, diabetic rats treated with YLE. Type 1 diabetes (T1DM) was induced by the administration of streptozotocin (STZ; 40 mg−1/kg body weight, single dose, i.p.), and treated groups received 100 mg/kg body weight YLE daily via gavage for 30 d. The YLE group shows an improvement in dysmetabolism and cardiomyopathy in the diabetic condition (DM versus DM + Y) promoting a significant reduction of glycemia by 63.39%, an increase in insulin concentration by 49.30%, and a decrease in serum triacylglycerol and fatty acid contents by 0.39- and 0.43-fold, respectively, by ameliorating the pancreatic islet injury, as well as increasing the activity of the antioxidant enzymes (catalase, superoxide dismutase, and glutathione peroxidase) and decreasing the fibrosis and cellular disorganization in cardiac tissue. The apparent benefits of YLE seem to be mediated by ameliorating dysmetabolism and oxidative stress in pancreatic and cardiac tissues.
Collapse
|
133
|
Goldberg IJ. 2017 George Lyman Duff Memorial Lecture: Fat in the Blood, Fat in the Artery, Fat in the Heart: Triglyceride in Physiology and Disease. Arterioscler Thromb Vasc Biol 2018; 38:700-706. [PMID: 29419410 PMCID: PMC5864527 DOI: 10.1161/atvbaha.117.309666] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/10/2018] [Indexed: 12/31/2022]
Abstract
Cholesterol is not the only lipid that causes heart disease. Triglyceride supplies the heart and skeletal muscles with highly efficient fuel and allows for the storage of excess calories in adipose tissue. Failure to transport, acquire, and use triglyceride leads to energy deficiency and even death. However, overabundance of triglyceride can damage and impair tissues. Circulating lipoprotein-associated triglycerides are lipolyzed by lipoprotein lipase (LpL) and hepatic triglyceride lipase. We inhibited these enzymes and showed that LpL inhibition reduces high-density lipoprotein cholesterol by >50%, and hepatic triglyceride lipase inhibition shifts low-density lipoprotein to larger, more buoyant particles. Genetic variations that reduce LpL activity correlate with increased cardiovascular risk. In contrast, macrophage LpL deficiency reduces macrophage function and atherosclerosis. Therefore, muscle and macrophage LpL have opposite effects on atherosclerosis. With models of atherosclerosis regression that we used to study diabetes mellitus, we are now examining whether triglyceride-rich lipoproteins or their hydrolysis by LpL affect the biology of established plaques. Following our focus on triglyceride metabolism led us to show that heart-specific LpL hydrolysis of triglyceride allows optimal supply of fatty acids to the heart. In contrast, cardiomyocyte LpL overexpression and excess lipid uptake cause lipotoxic heart failure. We are now studying whether interrupting pathways for lipid uptake might prevent or treat some forms of heart failure.
Collapse
Affiliation(s)
- Ira J Goldberg
- From the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University School of Medicine.
| |
Collapse
|
134
|
Zhang Y, Li Y, Liu Z, Zhao Q, Zhang H, Wang X, Lu P, Yu H, Wang M, Dong H, Zhang Z. PEDF regulates lipid metabolism and reduces apoptosis in hypoxic H9c2 cells by inducing autophagy related 5-mediated autophagy via PEDF-R. Mol Med Rep 2018; 17:7170-7176. [PMID: 29568944 PMCID: PMC5928674 DOI: 10.3892/mmr.2018.8733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 11/16/2017] [Indexed: 01/15/2023] Open
Abstract
Pigment epithelial-derived factor (PEDF) is a multifunctional secreted glycoprotein, which exerts a variety of physiological activities. PEDF may protect against hypoxia‑induced cell death associated with its antioxidative effects and p53 mitochondrial translocation in cultured cardiomyocytes and H9c2 cells. Additionally, previous studies have suggested that autophagy is an important cell survival mechanism. However, the effect of PEDF on autophagy and the associated pathway in hypoxic H9c2 cells has not been fully established. Autophagy has been reported to regulate lipid metabolism; however, little is known about whether PEDF is able to regulate lipid metabolism by promoting autophagy. In the present study, western blotting results revealed that PEDF increased the level of microtubule‑associated protein 1A/1B‑light chain 3 (LC3)‑II. Transmission electron microscopy (TEM) and LC3 fluorescence demonstrated that PEDF increased the number of autophagosomes. PEDF also increased the viability of hypoxic H9c2 cells and decreased the level of cleaved caspase‑3 protein, as evidenced by CCK‑8 assays and western blotting, respectively. TEM and a triglyceride assay kit demonstrated that PEDF‑induced autophagy may stimulate lipid degradation. Western blotting results revealed a novel mechanism underlying PEDF‑induced H9c2 cell autophagy via the PEDF‑R‑mediated Atg5 pathway under hypoxic conditions. Furthermore, the results also suggest that PEDF‑induced autophagy may stimulate lipid degradation. The survival function of autophagy suggests that modulation of PEDF‑induced autophagy may be used as a therapeutic strategy to protect cells against lipid-associated metabolic diseases.
Collapse
Affiliation(s)
- Yiqian Zhang
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yufeng Li
- Department of Thoracic Cardiovascular Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Zhiwei Liu
- Research Facility Center for Morphology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Qixiang Zhao
- Department of Thoracic Cardiovascular Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Hao Zhang
- Department of Thoracic Cardiovascular Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Xiaoyu Wang
- Department of Thoracic Cardiovascular Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Peng Lu
- Department of Thoracic Cardiovascular Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Hongli Yu
- Research Facility Center for Morphology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Meng Wang
- Research Facility Center for Morphology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Hongyan Dong
- Research Facility Center for Morphology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Zhongming Zhang
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
135
|
Lushnikova EL, Semenov DE, Nikityuk DB, Koldysheva EV, Klinnikova MG. Intracellular Reorganization of Cardiomyocytes in Dyslipidemic Cardiomyopathies. Bull Exp Biol Med 2018; 164:508-513. [DOI: 10.1007/s10517-018-4022-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Indexed: 01/18/2023]
|
136
|
Jung Y, Park J, Kim HL, Sim JE, Youn DH, Kang J, Lim S, Jeong MY, Yang WM, Lee SG, Ahn KS, Um JY. Vanillic acid attenuates obesity via activation of the AMPK pathway and thermogenic factors in vivo and in vitro. FASEB J 2018; 32:1388-1402. [PMID: 29141998 DOI: 10.1096/fj.201700231rr] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Energy expenditure is a target gaining recent interest for obesity treatment. The antiobesity effect of vanillic acid (VA), a well-known flavoring agent, was investigated in vivo and in vitro. High-fat diet (HFD)-induced obese mice and genetically obese db/db mice showed significantly decreased body weights after VA administration. Two major adipogenic markers, peroxisome proliferator activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα), were reduced while the key factor of energy metabolism, AMPKα, was increased in the white adipose tissue and liver tissue of VA-treated mice. Furthermore, VA inhibited lipid accumulation and reduced hepatotoxic/inflammatory markers in liver tissues of mice and HepG2 hepatocytes. VA treatment also decreased differentiation of 3T3-L1 adipocytes by regulating adipogenic factors including PPARγ and C/EBPα. AMPKα small interfering RNA was used to examine whether AMPK was associated with the actions of VA. In AMPKα-nulled 3T3-L1 cells, the inhibitory action of VA on PPARγ and C/EBPα was attenuated. Furthermore, in brown adipose tissues of mice and primary cultured brown adipocytes, VA increased mitochondria- and thermogenesis-related factors such as uncoupling protein 1 and PPARγ-coactivator 1-α. Taken together, our results suggest that VA has potential as an AMPKα- and thermogenesis-activating antiobesity agent.-Jung, Y., Park, J., Kim, H.-L., Sim, J.-E., Youn, D.-H., Kang, J., Lim, S., Jeong, M.-Y., Yang, W. M., Lee, S.-G., Ahn, K. S., Um, J.-Y. Vanillic acid attenuates obesity via activation of the AMPK pathway and thermogenic factors in vivo and in vitro.
Collapse
Affiliation(s)
- Yunu Jung
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Jinbong Park
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Hye-Lin Kim
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Jung-Eun Sim
- Department of Biological Sciences in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Dong-Hyun Youn
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - JongWook Kang
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Seona Lim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Mi-Young Jeong
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Woong Mo Yang
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Seok-Geun Lee
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Kwang Seok Ahn
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Jae-Young Um
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
137
|
Liu Y, Conlon DM, Bi X, Slovik KJ, Shi J, Edelstein HI, Millar JS, Javaheri A, Cuchel M, Pashos EE, Iqbal J, Hussain MM, Hegele RA, Yang W, Duncan SA, Rader DJ, Morrisey EE. Lack of MTTP Activity in Pluripotent Stem Cell-Derived Hepatocytes and Cardiomyocytes Abolishes apoB Secretion and Increases Cell Stress. Cell Rep 2018; 19:1456-1466. [PMID: 28514664 DOI: 10.1016/j.celrep.2017.04.064] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 02/22/2017] [Accepted: 04/21/2017] [Indexed: 01/26/2023] Open
Abstract
Abetalipoproteinemia (ABL) is an inherited disorder of lipoprotein metabolism resulting from mutations in microsomal triglyceride transfer protein (MTTP). In addition to expression in the liver and intestine, MTTP is expressed in cardiomyocytes, and cardiomyopathy has been reported in several ABL cases. Using induced pluripotent stem cells (iPSCs) generated from an ABL patient homozygous for a missense mutation (MTTPR46G), we show that human hepatocytes and cardiomyocytes exhibit defects associated with ABL disease, including loss of apolipoprotein B (apoB) secretion and intracellular accumulation of lipids. MTTPR46G iPSC-derived cardiomyocytes failed to secrete apoB, accumulated intracellular lipids, and displayed increased cell death, suggesting intrinsic defects in lipid metabolism due to loss of MTTP function. Importantly, these phenotypes were reversed after the correction of the MTTPR46G mutation by CRISPR/Cas9 gene editing. Together, these data reveal clear cellular defects in iPSC-derived hepatocytes and cardiomyocytes lacking MTTP activity, including a cardiomyocyte-specific regulated stress response to elevated lipids.
Collapse
Affiliation(s)
- Ying Liu
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Donna M Conlon
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xin Bi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine J Slovik
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jianting Shi
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hailey I Edelstein
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John S Millar
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ali Javaheri
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marina Cuchel
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Evanthia E Pashos
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jahangir Iqbal
- Department of Cell Biology and Pediatrics, State University of New York Downstate Medicine Center, Brooklyn, NY 11203, USA
| | - M Mahmood Hussain
- Department of Cell Biology and Pediatrics, State University of New York Downstate Medicine Center, Brooklyn, NY 11203, USA
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Wenli Yang
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen A Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Daniel J Rader
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Edward E Morrisey
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
138
|
Law BA, Liao X, Moore KS, Southard A, Roddy P, Ji R, Szulc Z, Bielawska A, Schulze PC, Cowart LA. Lipotoxic very-long-chain ceramides cause mitochondrial dysfunction, oxidative stress, and cell death in cardiomyocytes. FASEB J 2018; 32:1403-1416. [PMID: 29127192 DOI: 10.1096/fj.201700300r] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Accumulating data support a role for bioactive lipids as mediators of lipotixicity in cardiomyocytes. One class of these, the ceramides, constitutes a family of molecules that differ in structure and are synthesized by distinct enzymes, ceramide synthase (CerS)1-CerS6. Data support that specific ceramides and the enzymes that catalyze their formation play distinct roles in cell function. In a mouse model of diabetic cardiomyopathy, sphingolipid profiling revealed increases in not only the CerS5-derived ceramides but also in very long chain (VLC) ceramides derived from CerS2. Overexpression of CerS2 elevated VLC ceramides caused insulin resistance, oxidative stress, mitochondrial dysfunction, and mitophagy. Palmitate induced CerS2 and oxidative stress, mitophagy, and apoptosis, which were prevented by depletion of CerS2. Neither overexpression nor knockdown of CerS5 had any function in these processes, suggesting a chain-length dependent impact of ceramides on mitochondrial function. This concept was also supported by the observation that synthetic mitochondria-targeted ceramides led to mitophagy in a manner proportional to N-acyl chain length. Finally, blocking mitophagy exacerbated cell death. Taken together, our results support a model by which CerS2 and VLC ceramides have a distinct role in lipotoxicity, leading to mitochondrial damage, which results in subsequent adaptive mitophagy. Our data reveal a novel lipotoxic pathway through CerS2.-Law, B. A., Liao, X., Moore, K. S., Southard, A., Roddy, P., Ji, R., Szulc, Z., Bielawska, A., Schulze, P. C., Cowart, L. A. Lipotoxic very-long-chain ceramides cause mitochondrial dysfunction, oxidative stress, and cell death in cardiomyocytes.
Collapse
Affiliation(s)
- Brittany A Law
- Department of Medicine-Cardiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Xianghai Liao
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Kelsey S Moore
- Department of Biochemistry and Molecular Biology, The Medical University of South Carolina, Charleston, South Carolina, USA
| | - Abigail Southard
- Department of Biochemistry and Molecular Biology, The Medical University of South Carolina, Charleston, South Carolina, USA
| | - Patrick Roddy
- Department of Biochemistry and Molecular Biology, The Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ruiping Ji
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Zdzislaw Szulc
- Department of Biochemistry and Molecular Biology, The Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ala Bielawska
- Department of Biochemistry and Molecular Biology, The Medical University of South Carolina, Charleston, South Carolina, USA
| | - P Christian Schulze
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, New York, USA.,Division of Cardiology, Angiology, Pneumology, and Intensive Medical Care, Department of Internal Medicine I, Friedrich-Schiller-University Jena, University of Jena, Jena, Germany; and
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology, The Medical University of South Carolina, Charleston, South Carolina, USA.,Department of Veteran's Affairs, Charleston, South Carolina, USA
| |
Collapse
|
139
|
Chen Z, Li Y, Wang Y, Qian J, Ma H, Wang X, Jiang G, Liu M, An Y, Ma L, Kang L, Jia J, Yang C, Zhang G, Chen Y, Gao W, Fu M, Huang Z, Tang H, Zhu Y, Ge J, Gong H, Zou Y. Cardiomyocyte-Restricted Low Density Lipoprotein Receptor-Related Protein 6 (LRP6) Deletion Leads to Lethal Dilated Cardiomyopathy Partly Through Drp1 Signaling. Am J Cancer Res 2018; 8:627-643. [PMID: 29344294 PMCID: PMC5771081 DOI: 10.7150/thno.22177] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/14/2017] [Indexed: 11/17/2022] Open
Abstract
Low density lipoprotein receptor-related protein 6 (LRP6), a wnt co-receptor, regulates multiple functions in various organs. However, the roles of LRP6 in the adult heart are not well understood. Methods: We observed LRP6 expression in heart with end-stage dilated cardiomyopathy (DCM) by western blot. Tamoxifen-inducible cardiac-specific LRP6 knockout mouse was constructed. Hemodynamic and echocardiographic analyses were performed to these mice. Results: Cardiac LRP6 expression was dramatically decreased in patients with end-stage dilated cardiomyopathy (DCM) compared to control group. Tamoxifen-inducible cardiac-specific LRP6 knockout mice developed acute heart failure and mitochondrial dysfunction with reduced survival. Proteomic analysis suggests the fatty acid metabolism disorder involving peroxisome proliferator-activated receptors (PPARs) signaling in the LRP6 deficient heart. Accumulation of mitochondrial targeting to autophagosomes and lipid droplet were observed in LRP6 deletion hearts. Further analysis revealed cardiac LRP6 deletion suppressed autophagic degradation and fatty acid utilization, coinciding with activation of dynamin-related protein 1 (Drp1) and downregulation of nuclear TFEB (Transcription factor EB). Injection of Mdivi-1, a Drp1 inhibitor, not only promoted nuclear translocation of TFEB, but also partially rescued autophagic degradation, improved PPARs signaling, and attenuated cardiac dysfunction induced by cardiac specific LRP6 deletion. Conclusions: Cardiac LRP6 deficiency greatly suppressed autophagic degradation and fatty acid utilization, and subsequently leads to lethal dilated cardiomyopathy and cardiac dysfunction through activation of Drp1 signaling. It suggests that heart failure progression may be attenuated by therapeutic modulation of LRP6 expression.
Collapse
|
140
|
Effect of Gegen Qinlian Decoction on Cardiac Gene Expression in Diabetic Mice. Int J Genomics 2017; 2017:7421761. [PMID: 29379793 PMCID: PMC5742884 DOI: 10.1155/2017/7421761] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/30/2017] [Accepted: 10/26/2017] [Indexed: 02/07/2023] Open
Abstract
The aim of this research is to investigate the therapeutic effect of GGQL decoction on cardiac dysfunction and elucidate the pharmacological mechanisms. db/db mice were divided into DB group or GGQL group, and WT mice were used as control. All mice were accessed by echocardiography. And the total RNA of LV tissue samples was sequenced, then differential expression genes were analyzed. The RNA-seq results were validated by the results of RT-qPCR of 4 genes identified as differentially expressed. The content of pyruvate and ceramide in myocardial tissue was also measured. The results showed that GGQL decoction could significantly improve the diastolic dysfunction, increase the content of pyruvate, and had the trend to reduce the ceramide content. The results of RNA-seq showed that 2958 genes were differentially expressed when comparing the DB group with the WT group. Among them, compared with the DB group, 26 genes were differentially regulated in the GGQL group. The expression results of 4 genes were consistent with the RNA-seq results. Our study reveals that GGQL decoction has a therapeutic effect on diastolic dysfunction of the left ventricular and the effect may be related to its role in promoting myocardial glycolysis and decreasing the content of ceramide.
Collapse
|
141
|
Singh AK, Raj V, Keshari AK, Rai A, Kumar P, Rawat A, Maity B, Kumar D, Prakash A, De A, Samanta A, Bhattacharya B, Saha S. Isolated mangiferin and naringenin exert antidiabetic effect via PPAR γ/GLUT4 dual agonistic action with strong metabolic regulation. Chem Biol Interact 2017; 280:33-44. [PMID: 29223569 DOI: 10.1016/j.cbi.2017.12.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/07/2017] [Accepted: 12/01/2017] [Indexed: 01/12/2023]
Abstract
In this study, we isolated two compounds from the leaves of Salacia oblonga (SA1, mangiferin and SA2, naringenin), and their structures were confirmed by infrared spectroscopy, nuclear magnetic resonance (NMR) spectroscopy, and mass spectrometry. SA1 and SA2 were orally administered to streptozotocin-induced diabetic rats at 50 and 100 mg/kg daily for 15 days. Blood glucose level, serum lipid profile, oxidative stress parameters, histopathology, docking, molecular parameters, and NMR-based metabolic perturbation studies were performed to investigate the pharmacological activities of SA1 and SA2. Results suggested that both compounds reduced blood glucose level, restored body weight, and normalized lipid concentrations in the serum and oxidative stress biomarkers in the liver and pancreas. In addition, the docking study on several diabetes-associated targets revealed that both compounds had a strong binding affinity towards peroxisome proliferator-activated receptor gamma (PPARγ) and glucose transporter type 4 (GLUT4). Further real-time reverse transcription polymerase chain reaction and western blot analyses were performed to confirm the gene and protein expression levels of PPARγ and GLUT4 in the pancreatic tissues. Data obtained from the molecular studies showed that both compounds exhibited antidiabetic effects through dual activation of PPARγ/GLUT4 signaling pathways. Finally, the NMR-based metabolic studies showed that both compounds normalized the diabetogenic metabolites in the serum. Altogether, we concluded that SA1 and SA2 might be potential antidiabetic lead compounds for future drug development.
Collapse
Affiliation(s)
- Ashok K Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, Uttar Pradesh, India
| | - Vinit Raj
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, Uttar Pradesh, India
| | - Amit K Keshari
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, Uttar Pradesh, India
| | - Amit Rai
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, Uttar Pradesh, India
| | - Pranesh Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, Uttar Pradesh, India
| | - Atul Rawat
- Centre of Biomedical Research (CBMR), Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India; Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Biswanath Maity
- Centre of Biomedical Research (CBMR), Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| | - Dinesh Kumar
- Centre of Biomedical Research (CBMR), Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| | - Anand Prakash
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Arnab De
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Amalesh Samanta
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Bolay Bhattacharya
- Geethanjali College of Pharmacy, Cheeryal, Keesara, Hyderabad 501301, India
| | - Sudipta Saha
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, Uttar Pradesh, India.
| |
Collapse
|
142
|
Ueno M, Suzuki J, Hirose M, Sato S, Imagawa M, Zenimaru Y, Takahashi S, Ikuyama S, Koizumi T, Konoshita T, Kraemer FB, Ishizuka T. Cardiac overexpression of perilipin 2 induces dynamic steatosis: prevention by hormone-sensitive lipase. Am J Physiol Endocrinol Metab 2017; 313:E699-E709. [PMID: 28851734 PMCID: PMC6415650 DOI: 10.1152/ajpendo.00098.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/24/2017] [Accepted: 08/24/2017] [Indexed: 11/22/2022]
Abstract
Cardiac intracellular lipid accumulation (steatosis) is a pathophysiological phenomenon observed in starvation and diabetes mellitus. Perilipin 2 (PLIN2) is a lipid droplet (LD)-associated protein expressed in nonadipose tissues, including the heart. To explore the pathophysiological function of myocardial PLIN2, we generated transgenic (Tg) mice by cardiac-specific overexpression of PLIN2. Tg hearts showed accumulation of numerous small LDs associated with mitochondrial chains and high cardiac triacylglycerol (TAG) content [8-fold greater than wild-type (WT) mice]. Despite massive steatosis, cardiac uptake of glucose, fatty acids and VLDL, systolic function, and expression of metabolic genes were comparable in the two genotypes, and no morphological changes were observed by electron microscopy in the Tg hearts. Twenty-four hours of fasting markedly reduced steatosis in Tg hearts, whereas WT mice showed accumulation of LDs. Although activity of adipose triglyceride lipase in heart homogenate was comparable between WT and Tg mice, activity of hormone-sensitive lipase (HSL) was 40-50% less in Tg than WT mice under both feeding and fasting conditions, suggesting interference of PLIN2 with HSL. Mice generated through crossing of PLIN2-Tg mice and HSL-Tg mice showed cardiac-specific HSL overexpression and complete lack of steatosis. The results suggest that cardiac PLIN2 plays an important pathophysiological role in the development of dynamic steatosis and that the latter was prevented by upregulation of intracellular lipases, including HSL.
Collapse
Affiliation(s)
- Masami Ueno
- Third Department of Internal Medicine, University of Fukui, Faculty of Medical Sciences, Fukui, Japan
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California; and
- Division of Endocrinology, Stanford University, Stanford, California
| | - Jinya Suzuki
- Third Department of Internal Medicine, University of Fukui, Faculty of Medical Sciences, Fukui, Japan;
| | | | - Satsuki Sato
- Third Department of Internal Medicine, University of Fukui, Faculty of Medical Sciences, Fukui, Japan
| | - Michiko Imagawa
- Third Department of Internal Medicine, University of Fukui, Faculty of Medical Sciences, Fukui, Japan
| | - Yasuo Zenimaru
- Third Department of Internal Medicine, University of Fukui, Faculty of Medical Sciences, Fukui, Japan
| | - Sadao Takahashi
- Division of Diabetes Medicine, Ageo Central General Hospital, Saitama, Japan
| | - Shoichiro Ikuyama
- Division of Endocrinology and Metabolism, Oita San-ai Medical Center, Oita, Japan
| | - Tsutomu Koizumi
- Research and Education Program for Life Science, University of Fukui, Faculty of Medical Sciences, Fukui, Japan
| | - Tadashi Konoshita
- Third Department of Internal Medicine, University of Fukui, Faculty of Medical Sciences, Fukui, Japan
| | - Fredric B Kraemer
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California; and
- Division of Endocrinology, Stanford University, Stanford, California
| | - Tamotsu Ishizuka
- Third Department of Internal Medicine, University of Fukui, Faculty of Medical Sciences, Fukui, Japan
| |
Collapse
|
143
|
Abstract
Angiogenesis plays an important role in controlling tissue development and maintaining normal tissue function. Dysregulated angiogenesis is implicated in the pathogenesis of a variety of diseases, particularly diabetes, cancers, and neurodegenerative disorders. As the major regulator of angiogenesis, the vascular endothelial growth factor (VEGF) family is composed of a group of crucial members including VEGF-B. While the physiological roles of VEGF-B remain debatable, increasing evidence suggests that this protein is able to protect certain type of cells from apoptosis under pathological conditions. More importantly, recent studies reveal that VEGF-B is involved in lipid transport and energy metabolism, implicating this protein in obesity, diabetes and related metabolic complications. This article summarizes the current knowledge and understanding of VEGF-B in physiology and pathology, and shed light on the therapeutic potential of this crucial protein.
Collapse
Affiliation(s)
- Hongyu Zhu
- a State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University , Nanjing , China
| | - Mingming Gao
- b Department of Pharmaceutical and Biomedical Sciences , University of Georgia , Athens , GA , USA
| | - Xiangdong Gao
- a State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University , Nanjing , China
| | - Yue Tong
- a State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University , Nanjing , China
| |
Collapse
|
144
|
Tsushima K, Bugger H, Wende AR, Soto J, Jenson GA, Tor AR, McGlauflin R, Kenny HC, Zhang Y, Souvenir R, Hu XX, Sloan CL, Pereira RO, Lira VA, Spitzer KW, Sharp TL, Shoghi KI, Sparagna GC, Rog-Zielinska EA, Kohl P, Khalimonchuk O, Schaffer JE, Abel ED. Mitochondrial Reactive Oxygen Species in Lipotoxic Hearts Induce Post-Translational Modifications of AKAP121, DRP1, and OPA1 That Promote Mitochondrial Fission. Circ Res 2017; 122:58-73. [PMID: 29092894 DOI: 10.1161/circresaha.117.311307] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 10/25/2017] [Accepted: 10/31/2017] [Indexed: 12/20/2022]
Abstract
RATIONALE Cardiac lipotoxicity, characterized by increased uptake, oxidation, and accumulation of lipid intermediates, contributes to cardiac dysfunction in obesity and diabetes mellitus. However, mechanisms linking lipid overload and mitochondrial dysfunction are incompletely understood. OBJECTIVE To elucidate the mechanisms for mitochondrial adaptations to lipid overload in postnatal hearts in vivo. METHODS AND RESULTS Using a transgenic mouse model of cardiac lipotoxicity overexpressing ACSL1 (long-chain acyl-CoA synthetase 1) in cardiomyocytes, we show that modestly increased myocardial fatty acid uptake leads to mitochondrial structural remodeling with significant reduction in minimum diameter. This is associated with increased palmitoyl-carnitine oxidation and increased reactive oxygen species (ROS) generation in isolated mitochondria. Mitochondrial morphological changes and elevated ROS generation are also observed in palmitate-treated neonatal rat ventricular cardiomyocytes. Palmitate exposure to neonatal rat ventricular cardiomyocytes initially activates mitochondrial respiration, coupled with increased mitochondrial polarization and ATP synthesis. However, long-term exposure to palmitate (>8 hours) enhances ROS generation, which is accompanied by loss of the mitochondrial reticulum and a pattern suggesting increased mitochondrial fission. Mechanistically, lipid-induced changes in mitochondrial redox status increased mitochondrial fission by increased ubiquitination of AKAP121 (A-kinase anchor protein 121) leading to reduced phosphorylation of DRP1 (dynamin-related protein 1) at Ser637 and altered proteolytic processing of OPA1 (optic atrophy 1). Scavenging mitochondrial ROS restored mitochondrial morphology in vivo and in vitro. CONCLUSIONS Our results reveal a molecular mechanism by which lipid overload-induced mitochondrial ROS generation causes mitochondrial dysfunction by inducing post-translational modifications of mitochondrial proteins that regulate mitochondrial dynamics. These findings provide a novel mechanism for mitochondrial dysfunction in lipotoxic cardiomyopathy.
Collapse
Affiliation(s)
- Kensuke Tsushima
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Heiko Bugger
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Adam R Wende
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Jamie Soto
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Gregory A Jenson
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Austin R Tor
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Rose McGlauflin
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Helena C Kenny
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Yuan Zhang
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Rhonda Souvenir
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Xiao X Hu
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Crystal L Sloan
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Renata O Pereira
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Vitor A Lira
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Kenneth W Spitzer
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Terry L Sharp
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Kooresh I Shoghi
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Genevieve C Sparagna
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Eva A Rog-Zielinska
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Peter Kohl
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Oleh Khalimonchuk
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Jean E Schaffer
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - E Dale Abel
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.).
| |
Collapse
|
145
|
Brown adipose tissue and lipid metabolism imaging. Methods 2017; 130:105-113. [DOI: 10.1016/j.ymeth.2017.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 04/28/2017] [Accepted: 05/05/2017] [Indexed: 01/20/2023] Open
|
146
|
Hu X, Xiao RP. MG53 and disordered metabolism in striated muscle. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1984-1990. [PMID: 29017896 DOI: 10.1016/j.bbadis.2017.10.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/06/2017] [Accepted: 10/06/2017] [Indexed: 12/25/2022]
Abstract
MG53 is a member of tripartite motif family (TRIM) that expressed most abundantly in striated muscle. Using rodent models, many studies have demonstrated the MG53 not only facilitates membrane repair after ischemia reperfusion injury, but also contributes to the protective effects of both pre- and post-conditioning. Recently, however, it has been shown that MG53 participates in the regulation of many metabolic processes, especially insulin signaling pathway. Thus, sustained overexpression of MG53 may contribute to the development of various metabolic disorders in striated muscle. In this review, using cardiac muscle as an example, we will discuss muscle metabolic disturbances associated with diabetes and the current understanding of the underlying molecular mechanisms; in particular, the pathogenesis of diabetic cardiomyopathy. We will focus on the pathways that MG53 regulates and how the dysregulation of MG53 leads to metabolic disorders, thereby establishing a causal relationship between sustained upregulation of MG53 and the development of muscle insulin resistance and metabolic disorders. 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.
Collapse
Affiliation(s)
- Xinli Hu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Beijing City Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Rui-Ping Xiao
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Beijing City Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China.
| |
Collapse
|
147
|
Imaging and lipidomics methods for lipid analysis in metabolic and cardiovascular disease. J Dev Orig Health Dis 2017; 8:566-574. [PMID: 28697812 DOI: 10.1017/s2040174417000496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiometabolic diseases exhibit changes in lipid biology, which is important as lipids have critical roles in membrane architecture, signalling, hormone synthesis, homoeostasis and metabolism. However, Developmental Origins of Health and Disease studies of cardiometabolic disease rarely include analysis of lipids. This short review highlights some examples of lipid pathology and then explores the technology available for analysing lipids, focussing on the need to develop imaging modalities for intracellular lipids. Analytical methods for studying interactions between the complex endocrine and intracellular signalling pathways that regulate lipid metabolism have been critical in expanding our understanding of how cardiometabolic diseases develop in association with obesity and dietary factors. Biochemical methods can be used to generate detailed lipid profiles to establish links between lifestyle factors and metabolic signalling pathways and determine how changes in specific lipid subtypes in plasma and homogenized tissue are associated with disease progression. New imaging modalities enable the specific visualization of intracellular lipid traffic and distribution in situ. These techniques provide a dynamic picture of the interactions between lipid storage, mobilization and signalling, which operate during normal cell function and are altered in many important diseases. The development of methods for imaging intracellular lipids can provide a dynamic real-time picture of how lipids are involved in complex signalling and other cell biology pathways; and how they ultimately regulate metabolic function/homoeostasis during early development. Some imaging modalities have the potential to be adapted for in vivo applications, and may enable the direct visualization of progression of pathogenesis of cardiometabolic disease after poor growth in early life.
Collapse
|
148
|
Talati MH, Brittain EL, Fessel JP, Penner N, Atkinson J, Funke M, Grueter C, Jerome WG, Freeman M, Newman JH, West J, Hemnes AR. Mechanisms of Lipid Accumulation in the Bone Morphogenetic Protein Receptor Type 2 Mutant Right Ventricle. Am J Respir Crit Care Med 2017; 194:719-28. [PMID: 27077479 DOI: 10.1164/rccm.201507-1444oc] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
RATIONALE In heritable pulmonary arterial hypertension with germline mutation in the bone morphogenetic protein receptor type 2 (BMPR2) gene, right ventricle (RV) dysfunction is associated with RV lipotoxicity; however, the underlying mechanism for lipid accumulation is not known. OBJECTIVES We hypothesized that lipid accumulation in cardiomyocytes with BMPR2 mutation occurs owing to alterations in lipid transport and impaired fatty acid oxidation (FAO), which is exacerbated by a high-lipid (Western) diet (WD). METHODS We used a transgenic mouse model of pulmonary arterial hypertension with mutant BMPR2 and generated a cardiomyocyte cell line with BMPR2 mutation. Electron microscopy and metabolomic analysis were performed on mouse RVs. MEASUREMENTS AND MAIN RESULTS By metabolomics analysis, we found an increase in long-chain fatty acids in BMPR2 mutant mouse RVs compared with controls, which correlated with cardiac index. BMPR2-mutant cardiomyocytes had increased lipid compared with controls. Direct measurement of FAO in the WD-fed BMPR2-mutant RV showed impaired palmitate-linked oxygen consumption, and metabolomics analysis showed reduced indices of FAO. Using both mutant BMPR2 mouse RVs and cardiomyocytes, we found an increase in the uptake of (14)C-palmitate and fatty acid transporter CD36 that was further exacerbated by WD. CONCLUSIONS Taken together, our data suggest that impaired FAO and increased expression of the lipid transporter CD36 are key mechanisms underlying lipid deposition in the BMPR2-mutant RV, which are exacerbated in the presence of dietary lipids. These findings suggest important features leading to RV lipotoxicity in pulmonary arterial hypertension and may point to novel areas of therapeutic intervention.
Collapse
Affiliation(s)
- Megha H Talati
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Joshua P Fessel
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee.,3 Department of Pharmacology
| | - Niki Penner
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Mitch Funke
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - W Gray Jerome
- 4 Department of Pathology, Microbiology, and Immunology.,6 Department of Cancer Biology, and
| | - Michael Freeman
- 7 Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John H Newman
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James West
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Anna R Hemnes
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
149
|
Abstract
For more than half a century, metabolic perturbations have been explored in the failing myocardium, highlighting a reversion to a more fetal-like metabolic profile (characterized by depressed fatty acid oxidation and concomitant increased reliance on use of glucose). More recently, alterations in ketone body and amino acid/protein metabolism have been described during heart failure, as well as mitochondrial dysfunction and perturbed metabolic signaling (e.g., acetylation, O-GlcNAcylation). Although numerous mechanisms are likely involved, the current review provides recent advances regarding the metabolic origins of heart failure, and their potential contribution toward contractile dysfunction of the heart.
Collapse
|
150
|
Thapa D, Zhang M, Manning JR, Guimarães DA, Stoner MW, O'Doherty RM, Shiva S, Scott I. Acetylation of mitochondrial proteins by GCN5L1 promotes enhanced fatty acid oxidation in the heart. Am J Physiol Heart Circ Physiol 2017; 313:H265-H274. [PMID: 28526709 DOI: 10.1152/ajpheart.00752.2016] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 12/22/2022]
Abstract
Lysine acetylation is a reversible posttranslational modification and is particularly important in the regulation of mitochondrial metabolic enzymes. Acetylation uses acetyl-CoA derived from fuel metabolism as a cofactor, thereby linking nutrition to metabolic activity. In the present study, we investigated how mitochondrial acetylation status in the heart is controlled by food intake and how these changes affect mitochondrial metabolism. We found that there was a significant increase in cardiac mitochondrial protein acetylation in mice fed a long-term high-fat diet and that this change correlated with an increase in the abundance of the mitochondrial acetyltransferase-related protein GCN5L1. We showed that the acetylation status of several mitochondrial fatty acid oxidation enzymes (long-chain acyl-CoA dehydrogenase, short-chain acyl-CoA dehydrogenase, and hydroxyacyl-CoA dehydrogenase) and a pyruvate oxidation enzyme (pyruvate dehydrogenase) was significantly upregulated in high-fat diet-fed mice and that the increase in long-chain and short-chain acyl-CoA dehydrogenase acetylation correlated with increased enzymatic activity. Finally, we demonstrated that the acetylation of mitochondrial fatty acid oxidation proteins was decreased after GCN5L1 knockdown and that the reduced acetylation led to diminished fatty acid oxidation in cultured H9C2 cells. These data indicate that lysine acetylation promotes fatty acid oxidation in the heart and that this modification is regulated in part by the activity of GCN5L1.NEW & NOTEWORTHY Recent research has shown that acetylation of mitochondrial fatty acid oxidation enzymes has greatly contrasting effects on their activity in different tissues. Here, we provide new evidence that acetylation of cardiac mitochondrial fatty acid oxidation enzymes by GCN5L1 significantly upregulates their activity in diet-induced obese mice.
Collapse
Affiliation(s)
- Dharendra Thapa
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and.,Center for Metabolism and Mitochondrial Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Manling Zhang
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and.,Center for Metabolism and Mitochondrial Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Janet R Manning
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and.,Center for Metabolism and Mitochondrial Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Danielle A Guimarães
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and.,Center for Metabolism and Mitochondrial Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael W Stoner
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and.,Center for Metabolism and Mitochondrial Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert M O'Doherty
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Metabolism and Mitochondrial Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and.,Center for Metabolism and Mitochondrial Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Iain Scott
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; .,Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and.,Center for Metabolism and Mitochondrial Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|