Inhibition of the Cardiomyocyte-Specific Kinase TNNI3K Limits Oxidative Stress, Injury, and Adverse Remodeling in the Ischemic Heart

Blocking the activity of a cardiomyocyte-specific protein kinase with a small-molecule inhibitor reduces oxidative stress, myocyte death, and adverse remodeling in the ischemic heart. Blocking Cardiac Kinase Prevents Heart Damage Restoring blood flow after a heart attack is essential; yet, rapid reperfusion of blood can cause adverse effects on heart cells (cardiomyocytes) via oxidative damage, calcium overload, and inflammation. To limit these effects, Vagnozzi and colleagues developed an inhibitor that targets a cardiomyocyte-specific kinase called TNNI3K, which may be intimately involved in signaling events after ischemia (blockage of blood flow) and reperfusion. The authors first confirmed that TNNI3K is up-regulated in tissues from patients with heart failure who were undergoing transplant. Mice that overexpressed active TNNI3K had larger infarcts than those with an inactive form of the kinase, as well as worse ischemic injury and cardiomyocyte death. Conversely, deletion of Tnni3k reduced infarct size and prevented cardiomyocyte death in mice. From the human tissues, the kinase appeared to be limited to cardiomyocytes, which lends itself to targeted therapy. Vagnozzi et al. administered two different small-molecule inhibitors during reperfusion to mice with ischemic injury and observed a reduction in left ventricle dysfunction, progressive remodeling, and fibrosis (a hardening of the heart tissue). The authors believe that these functional benefits stem from a concomitant reduction in superoxide production, p38 activation, and infarct size. This inhibition strategy will need to be tested in a large-animal model before translation. If successful, it could find immediate application to patients with chronic ischemic cardiomyopathy, where recurrent ischemia is followed by reperfusion. Percutaneous coronary intervention is first-line therapy for acute coronary syndromes (ACS) but can promote cardiomyocyte death and cardiac dysfunction via reperfusion injury, a phenomenon driven in large part by oxidative stress. Therapies to limit this progression have proven elusive, with no major classes of new agents since the development of anti-platelets/anti-thrombotics. We report that cardiac troponin I–interacting kinase (TNNI3K), a cardiomyocyte-specific kinase, promotes ischemia/reperfusion injury, oxidative stress, and myocyte death. TNNI3K-mediated injury occurs through increased mitochondrial superoxide production and impaired mitochondrial function and is largely dependent on p38 mitogen-activated protein kinase (MAPK) activation. We developed a series of small-molecule TNNI3K inhibitors that reduce mitochondrial-derived superoxide generation, p38 activation, and infarct size when delivered at reperfusion to mimic clinical intervention. TNNI3K inhibition also preserves cardiac function and limits chronic adverse remodeling. Our findings demonstrate that TNNI3K modulates reperfusion injury in the ischemic heart and is a tractable therapeutic target for ACS. Pharmacologic TNNI3K inhibition would be cardiac-selective, preventing potential adverse effects of systemic kinase inhibition.

[1]  Andres Metspalu,et al.  Genome-wide analysis of BMI in adolescents and young adults reveals additional insight into the effects of genetic loci over the life course. , 2013, Human molecular genetics.

[2]  R. V. Vander Heide,et al.  Cardioprotection and Myocardial Reperfusion: Pitfalls to Clinical Application , 2013, Circulation research.

[3]  R. Hui,et al.  TNNI3K, a Cardiac-Specific Kinase, Promotes Physiological Cardiac Hypertrophy in Transgenic Mice , 2013, PloS one.

[4]  Hui Wang,et al.  TNNI3K is a novel mediator of myofilament function and phosphorylates cardiac troponin I , 2013, Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas.

[5]  D. Yellon,et al.  Myocardial ischemia-reperfusion injury: a neglected therapeutic target. , 2013, The Journal of clinical investigation.

[6]  D. Marchuk,et al.  Dissection of a Quantitative Trait Locus for PR Interval Duration Identifies Tnni3k as a Novel Modulator of Cardiac Conduction , 2012, PLoS genetics.

[7]  M. Marber,et al.  New therapeutic targets in cardiology: p38 alpha mitogen-activated protein kinase for ischemic heart disease. , 2012, Circulation.

[8]  D. Kass,et al.  Animal models of heart failure: a scientific statement from the American Heart Association. , 2012, Circulation research.

[9]  R. Kloner,et al.  An update on cardioprotection: a review of the latest adjunctive therapies to limit myocardial infarction size in clinical trials. , 2012, Journal of the American College of Cardiology.

[10]  Joseph T. Glessner,et al.  Role of BMI‐Associated Loci Identified in GWAS Meta‐Analyses in the Context of Common Childhood Obesity in European Americans , 2011, Obesity.

[11]  C. Baines How and When Do Myocytes Die During Ischemia and Reperfusion: The Late Phase , 2011, Journal of cardiovascular pharmacology and therapeutics.

[12]  Hong Wang,et al.  Hyperhomocysteinemia impairs endothelium-derived hyperpolarizing factor-mediated vasorelaxation in transgenic cystathionine beta synthase-deficient mice. , 2011, Blood.

[13]  S. Vidal,et al.  Quantitative Trait Locus Analysis, Pathway Analysis, and Consomic Mapping Show Genetic Variants of Tnni3k, Fpgt, or H28 Control Susceptibility to Viral Myocarditis , 2011, The Journal of Immunology.

[14]  Hui Wang,et al.  Adenovirus‐mediated overexpression of cardiac troponin I‐interacting kinase promotes cardiomyocyte hypertrophy , 2011, Clinical and experimental pharmacology & physiology.

[15]  W. Koch,et al.  A Novel and Efficient Model of Coronary Artery Ligation and Myocardial Infarction in the Mouse , 2010, Circulation research.

[16]  Yibin Wang,et al.  Mitogen-activated protein kinase signaling in the heart: angels versus demons in a heart-breaking tale. , 2010, Physiological reviews.

[17]  Joseph A. Hill,et al.  Pathogenesis of myocardial ischemia-reperfusion injury and rationale for therapy. , 2010, The American journal of cardiology.

[18]  J. Molkentin,et al.  Developing small molecules to inhibit kinases unkind to the heart: p38 MAPK as a case in point. , 2010, Drug discovery today. Disease mechanisms.

[19]  T. Hadnott,et al.  Tnni3k Modifies Disease Progression in Murine Models of Cardiomyopathy , 2009, PLoS genetics.

[20]  Y. Pinto,et al.  Avoidance of Transient Cardiomyopathy in Cardiomyocyte-Targeted Tamoxifen-Induced MerCreMer Gene Deletion Models , 2009, Circulation research.

[21]  C. Baines The mitochondrial permeability transition pore and ischemia-reperfusion injury , 2009, Basic Research in Cardiology.

[22]  I. Komuro,et al.  Overexpression of TNNI3K, a cardiac-specific MAP kinase, promotes P19CL6-derived cardiac myogenesis and prevents myocardial infarction-induced injury. , 2008, American Journal of Physiology. Heart and Circulatory Physiology.

[23]  E. Murphy,et al.  Mechanisms underlying acute protection from cardiac ischemia-reperfusion injury. , 2008, Physiological reviews.

[24]  D. Yellon,et al.  Myocardial reperfusion injury. , 2007, The New England journal of medicine.

[25]  J. Ding,et al.  Identification of the dual specificity and the functional domains of the cardiac-specific protein kinase TNNI3K. , 2007, General physiology and biophysics.

[26]  Jenine K Anday,et al.  Gene ancestry of the cannabinoid receptor family. , 2005, Pharmacological research.

[27]  Yow-Ming C Wang,et al.  c-Jun N-Terminal Kinases Mediate Reactivation of Akt and Cardiomyocyte Survival After Hypoxic Injury In Vitro and In Vivo , 2005, Circulation research.

[28]  Z. Ao,et al.  Role of p38 MAP kinase in postcapillary venule leukocyte adhesion induced by ischemia/reperfusion injury. , 2005, Pharmacological research.

[29]  L. Becker New concepts in reactive oxygen species and cardiovascular reperfusion physiology. , 2004, Cardiovascular research.

[30]  Peipei Ping,et al.  Role of the mitochondrial permeability transition in myocardial disease. , 2003, Circulation research.

[31]  Ying-jie Wei,et al.  Cloning and characterization of a novel cardiac-specific kinase that interacts specifically with cardiac troponin I , 2003, Journal of Molecular Medicine.

[32]  N. Chandel,et al.  Mitochondrial ROS initiate phosphorylation of p38 MAP kinase during hypoxia in cardiomyocytes. , 2002, American journal of physiology. Lung cellular and molecular physiology.

[33]  John C. Lee,et al.  Hypertensive End-Organ Damage and Premature Mortality Are p38 Mitogen-Activated Protein Kinase–Dependent in a Rat Model of Cardiac Hypertrophy and Dysfunction , 2001, Circulation.

[34]  D. Lefer,et al.  Oxidative stress and cardiac disease. , 2000, The American journal of medicine.

[35]  A. Clerk,et al.  Stimulation of “Stress-regulated” Mitogen-activated Protein Kinases (Stress-activated Protein Kinases/c-Jun N-terminal Kinases and p38-Mitogen-activated Protein Kinases) in Perfused Rat Hearts by Oxidative and Other Stresses* , 1998, The Journal of Biological Chemistry.

[36]  T. Vanden Hoek,et al.  Significant levels of oxidants are generated by isolated cardiomyocytes during ischemia prior to reperfusion. , 1997, Journal of molecular and cellular cardiology.

[37]  G. Ghai,et al.  Myocardial alterations due to free-radical generation. , 1984, The American journal of physiology.

[38]  D. Marchuk,et al.  Overexpression of TNNI3K, a cardiac-specific MAPKKK, promotes cardiac dysfunction. , 2013, Journal of molecular and cellular cardiology.

[39]  M. Marber,et al.  Basic Science for Clinicians New Therapeutic Targets in Cardiology p38 Alpha Mitogen-Activated Protein Kinase for Ischemic Heart Disease , 2012 .

[40]  Matthew Larkin,et al.  National Heart Lung and Blood Institute, National Institute of Health , 2012 .

[41]  S. Vatner,et al.  Inhibition of p38 alpha MAPK rescues cardiomyopathy induced by overexpressed beta 2-adrenergic receptor, but not beta 1-adrenergic receptor. , 2007, The Journal of clinical investigation.