ATF4 Protects the Heart From Failure by Antagonizing Oxidative Stress

Background: Cellular redox control is maintained by generation of reactive oxygen/nitrogen species balanced by activation of antioxidative pathways. Disruption of redox balance leads to oxidative stress, a central causative event in numerous diseases including heart failure. Redox control in the heart exposed to hemodynamic stress, however, remains to be fully elucidated. Methods: Pressure overload was triggered by transverse aortic constriction in mice. Transcriptomic and metabolomic regulations were evaluated by RNA-sequencing and metabolomics, respectively. Stable isotope tracer labeling experiments were conducted to determine metabolic flux in vitro. Neonatal rat ventricular myocytes and H9c2 cells were used to examine molecular mechanisms. Results: We show that production of cardiomyocyte NADPH, a key factor in redox regulation, is decreased in pressure overload-induced heart failure. As a consequence, the level of reduced glutathione is downregulated, a change associated with fibrosis and cardiomyopathy. We report that the pentose phosphate pathway and mitochondrial serine/glycine/folate metabolic signaling, 2 NADPH-generating pathways in the cytosol and mitochondria, respectively, are induced by transverse aortic constriction. We identify ATF4 (activating transcription factor 4) as an upstream transcription factor controlling the expression of multiple enzymes in these 2 pathways. Consistently, joint pathway analysis of transcriptomic and metabolomic data reveal that ATF4 preferably controls oxidative stress and redox-related pathways. Overexpression of ATF4 in neonatal rat ventricular myocytes increases NADPH-producing enzymes‚ whereas silencing of ATF4 decreases their expression. Further, stable isotope tracer experiments reveal that ATF4 overexpression augments metabolic flux within these 2 pathways. In vivo, cardiomyocyte-specific deletion of ATF4 exacerbates cardiomyopathy in the setting of transverse aortic constriction and accelerates heart failure development, attributable, at least in part, to an inability to increase the expression of NADPH-generating enzymes. Conclusions: Our findings reveal that ATF4 plays a critical role in the heart under conditions of hemodynamic stress by governing both cytosolic and mitochondrial production of NADPH.

[1]  Rizwan Kalani,et al.  Heart Disease and Stroke Statistics—2022 Update: A Report From the American Heart Association , 2022, Circulation.

[2]  J. Molkentin,et al.  Thbs1 induces lethal cardiac atrophy through PERK-ATF4 regulated autophagy , 2021, Nature communications.

[3]  M. V. Vander Heiden,et al.  PKM1 Exerts Critical Roles in Cardiac Remodeling Under Pressure Overload in the Heart , 2021, Circulation.

[4]  J. Xia,et al.  MetaboAnalyst 5.0: narrowing the gap between raw spectra and functional insights , 2021, Nucleic Acids Res..

[5]  R. Deberardinis,et al.  Mitochondrial NADP+ is essential for proline biosynthesis during cell growth , 2021, Nature Metabolism.

[6]  K. De Keersmaecker,et al.  The ins and outs of serine and glycine metabolism in cancer , 2021, Nature Metabolism.

[7]  K. Parnell,et al.  The pyruvate-lactate axis modulates cardiac hypertrophy and heart failure. , 2020, Cell metabolism.

[8]  M. Crespo-Leiro,et al.  Mitochondrial pyruvate carrier abundance mediates pathological cardiac hypertrophy , 2020, Nature Metabolism.

[9]  O. Ilkayeva,et al.  Nutritional modulation of heart failure in mitochondrial pyruvate carrier–deficient mice , 2020, Nature Metabolism.

[10]  D. Xie,et al.  NADPH homeostasis in cancer: functions, mechanisms and therapeutic implications , 2020, Signal Transduction and Targeted Therapy.

[11]  J. Asara,et al.  The mTORC1-mediated activation of ATF4 promotes protein and glutathione synthesis downstream of growth signals , 2020, bioRxiv.

[12]  Paul V. Taufalele,et al.  Mitochondrial Pyruvate Carriers are Required for Myocardial Stress Adaptation , 2020, Nature Metabolism.

[13]  M. Packer Uric Acid Is a Biomarker of Oxidative Stress in the Failing Heart: Lessons Learned from Trials With Allopurinol and SGLT2 Inhibitors. , 2020, Journal of cardiac failure.

[14]  Herman I. May,et al.  Chronic activation of hexosamine biosynthesis in the heart triggers pathological cardiac remodeling , 2020, Nature Communications.

[15]  G. Hart,et al.  Excessive O - GlcNAcylation causes heart failure and sudden death , 2020, bioRxiv.

[16]  Herman I. May,et al.  Spliced X-Box Binding Protein 1 Stimulates Adaptive Growth Through Activation of mTOR. , 2019, Circulation.

[17]  Z. Ignatova,et al.  ATF4 couples MYC-dependent translational activity to bioenergetic demands during tumor progression , 2019, Nature Cell Biology.

[18]  J. Weinstein,et al.  ElemCor: accurate data analysis and enrichment calculation for high-resolution LC-MS stable isotope labeling experiments , 2019, BMC Bioinformatics.

[19]  Xiang Luo,et al.  GRP78 (Glucose-Regulated Protein of 78 kDa) Promotes Cardiomyocyte Growth Through Activation of GATA4 (GATA-Binding Protein 4) , 2019, Hypertension.

[20]  B. Hill,et al.  Metabolic Coordination of Physiological and Pathological Cardiac Remodeling , 2018, Circulation research.

[21]  J. Loscalzo,et al.  NAD(H) and NADP(H) Redox Couples and Cellular Energy Metabolism. , 2018, Antioxidants & redox signaling.

[22]  B. Viollet,et al.  AMPK activation counteracts cardiac hypertrophy by reducing O-GlcNAcylation , 2018, Nature Communications.

[23]  O. Gorbatyuk,et al.  Neurodegeneration: Keeping ATF4 on a Tight Leash , 2017, Front. Cell. Neurosci..

[24]  Herman I. May,et al.  Temporal dynamics of cardiac hypertrophic growth in response to pressure overload. , 2017, American journal of physiology. Heart and circulatory physiology.

[25]  Inge M. N. Wortel,et al.  Surviving Stress: Modulation of ATF4-Mediated Stress Responses in Normal and Malignant Cells , 2017, Trends in Endocrinology & Metabolism.

[26]  E. Pearce,et al.  Ancillary Activity: Beyond Core Metabolism in Immune Cells. , 2017, Cell metabolism.

[27]  Joshua D Rabinowitz,et al.  One-Carbon Metabolism in Health and Disease. , 2017, Cell metabolism.

[28]  Gianluigi Savarese,et al.  Global Public Health Burden of Heart Failure. , 2016, Cardiac failure review.

[29]  J. Rabinowitz,et al.  Malic enzyme tracers reveal hypoxia-induced switch in adipocyte NADPH pathway usage. , 2016, Nature chemical biology.

[30]  Q. Wang,et al.  Fatty acid oxidation and carnitine palmitoyltransferase I: emerging therapeutic targets in cancer , 2016, Cell Death and Disease.

[31]  J. Asara,et al.  mTORC1 induces purine synthesis through control of the mitochondrial tetrahydrofolate cycle , 2016, Science.

[32]  G. Figtree,et al.  Redox biomarkers in cardiovascular medicine. , 2015, European heart journal.

[33]  N. Hay,et al.  The pentose phosphate pathway and cancer. , 2014, Trends in biochemical sciences.

[34]  T. Shlomi,et al.  Quantitative flux analysis reveals folate-dependent NADPH production , 2014, Nature.

[35]  P. Storz Oxidative Stress in Cancer , 2013 .

[36]  N. Hay,et al.  Molecular Pathways: Reactive Oxygen Species Homeostasis in Cancer Cells and Implications for Cancer Therapy , 2013, Clinical Cancer Research.

[37]  D. Kass,et al.  Nitric oxide synthases in heart failure. , 2013, Antioxidants & redox signaling.

[38]  I. Mills,et al.  ER stress-mediated autophagy promotes Myc-dependent transformation and tumor growth. , 2012, The Journal of clinical investigation.

[39]  Steven D. Kunkel,et al.  Stress-induced Skeletal Muscle Gadd45a Expression Reprograms Myonuclei and Causes Muscle Atrophy* , 2012, The Journal of Biological Chemistry.

[40]  P. Ray,et al.  Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. , 2012, Cellular signalling.

[41]  C. Palmeira,et al.  Role of oxidative stress in the pathogenesis of nonalcoholic steatohepatitis. , 2012, Free radical biology & medicine.

[42]  Hongqiao Zhang,et al.  Glutathione: overview of its protective roles, measurement, and biosynthesis. , 2009, Molecular aspects of medicine.

[43]  W. Ying NAD+/NADH and NADP+/NADPH in cellular functions and cell death: regulation and biological consequences. , 2008, Antioxidants & redox signaling.

[44]  D. Harrison,et al.  Molecular Mechanisms of Angiotensin II–Mediated Mitochondrial Dysfunction: Linking Mitochondrial Oxidative Damage and Vascular Endothelial Dysfunction , 2007, Circulation research.

[45]  Hiroyuki Tsutsui,et al.  [Oxidative stress and heart failure]. , 2006, Nihon rinsho. Japanese journal of clinical medicine.

[46]  A. Shah,et al.  Oxidative stress in heart failure. More than just damage. , 2003, European heart journal.

[47]  L. Dell’Italia,et al.  Cardiac interstitial bradykinin and mast cells modulate pattern of LV remodeling in volume overload in rats. , 2003, American journal of physiology. Heart and circulatory physiology.

[48]  A. Shah,et al.  Increased myocardial NADPH oxidase activity in human heart failure. , 2003, Journal of the American College of Cardiology.

[49]  D. Kass,et al.  Allopurinol Improves Myocardial Efficiency in Patients With Idiopathic Dilated Cardiomyopathy , 2001, Circulation.

[50]  J. Ross,et al.  Segregation of atrial-specific and inducible expression of an atrial natriuretic factor transgene in an in vivo murine model of cardiac hypertrophy , 1991, Proceedings of the National Academy of Sciences of the United States of America.