NLRX1 Deletion Increases Ischemia-Reperfusion Damage and Activates Glucose Metabolism in Mouse Heart

Background NOD-like receptors (NLR) are intracellular sensors of the innate immune system, with the NLRP3 being a pro-inflammatory member that modulates cardiac ischemia-reperfusion injury (IRI) and metabolism. No information is available on a possible role of anti-inflammatory NLRs on IRI and metabolism in the intact heart. Here we hypothesize that the constitutively expressed, anti-inflammatory mitochondrial NLRX1, affects IRI and metabolism of the isolated mouse heart. Methods Isolated C57Bl/6J and NLRX1 knock-out (KO) mouse hearts were perfused with a physiological mixture of the essential substrates (lactate, glucose, pyruvate, fatty acid, glutamine) and insulin. For the IRI studies, hearts were subjected to either mild (20 min) or severe (35 min) ischemia and IRI was determined at 60 min reperfusion. Inflammatory mediators (IL-6, TNFα) and survival pathways (mito-HKII, p-Akt, p-AMPK, p-STAT3) were analyzed at 5 min of reperfusion. For the metabolism studies, hearts were perfused for 35 min with either 5.5 mM 13C-glucose or 0.4 mM 13C-palmitate under normoxic conditions, followed by LC-MS analysis and integrated, stepwise, mass-isotopomeric flux analysis (MIMOSA). Results NLRX1 KO significantly increased IRI (infarct size from 63% to 73%, end-diastolic pressure from 59 mmHg to 75 mmHg, and rate-pressure-product recovery from 15% to 6%), following severe, but not mild, ischemia. The increased IRI in NLRX1 KO hearts was associated with depressed Akt signaling at early reperfusion; other survival pathways or inflammatory parameters were not affected. Metabolically, NLRX1 KO hearts displayed increased lactate production and glucose oxidation relative to fatty acid oxidation, associated with increased pyruvate dehydrogenase flux and 10% higher cardiac oxygen consumption. Conclusion Deletion of the mitochondrially-located NOD-like sensor NLRX1 exacerbates severe cardiac IR injury, possibly through impaired Akt signaling, and increases cardiac glucose metabolism.

[1]  Michael L. Blackburn,et al.  Carnitine palmitoyltransferase 2 (CPT2) knockout potentiates palmitate-induced insulin resistance in C2C12 myotubes. , 2020, American journal of physiology. Endocrinology and metabolism.

[2]  L. Bertrand,et al.  Cardiac metabolism as a driver and therapeutic target of myocardial infarction , 2020, Journal of cellular and molecular medicine.

[3]  J. Bassaganya-Riera,et al.  Activation of NLRX1 by NX-13 Alleviates Inflammatory Bowel Disease through Immunometabolic Mechanisms in CD4+ T Cells , 2019, The Journal of Immunology.

[4]  I. C. Allen,et al.  NLRX1 Is a Multifaceted and Enigmatic Regulator of Immune System Function , 2019, Front. Immunol..

[5]  P. Calabresi,et al.  NLRX1 inhibits the early stages of CNS inflammation and prevents the onset of spontaneous autoimmunity , 2019, PLoS biology.

[6]  R. Coronel,et al.  Delayed ischemic contracture onset by Empagliflozin associates with NHE-1 inhibition and is dependent on insulin in isolated mouse hearts. , 2019, Cardiovascular research.

[7]  J. Downey,et al.  Innate immunity as a target for acute cardioprotection. , 2019, Cardiovascular research.

[8]  P. Chumakov,et al.  NLRX1 regulates TNF-α-induced mitochondria-lysosomal crosstalk to maintain the invasive and metastatic potential of breast cancer cells. , 2019, Biochimica et biophysica acta. Molecular basis of disease.

[9]  R. Spang,et al.  Correcting for natural isotope abundance and tracer impurity in MS-, MS/MS- and high-resolution-multiple-tracer-data from stable isotope labeling experiments with IsoCorrectoR , 2018, Scientific Reports.

[10]  D. Philpott,et al.  The mitochondrial Nod-like receptor NLRX1 modifies apoptosis through SARM1 , 2018, Molecular and Cellular Biochemistry.

[11]  J. Auwerx,et al.  Increased cardiac fatty acid oxidation in a mouse model with decreased malonyl-CoA sensitivity of CPT1B , 2018, Cardiovascular research.

[12]  S. Denis,et al.  Pyruvate dehydrogenase complex plays a central role in brown adipocyte energy expenditure and fuel utilization during short-term beta-adrenergic activation , 2018, Scientific Reports.

[13]  S. Girardin,et al.  Deletion of NLRX1 increases fatty acid metabolism and prevents diet-induced hepatic steatosis and metabolic syndrome. , 2018, Biochimica et biophysica acta. Molecular basis of disease.

[14]  A. Abbate,et al.  The NLRP3 inflammasome in acute myocardial infarction , 2018, Nature Reviews Cardiology.

[15]  Xiao-dong Zhu,et al.  NOD-like receptor X1 functions as a tumor suppressor by inhibiting epithelial-mesenchymal transition and inducing aging in hepatocellular carcinoma cells , 2018, Journal of Hematology & Oncology.

[16]  E. Liepinsh,et al.  Acute and long‐term administration of palmitoylcarnitine induces muscle‐specific insulin resistance in mice , 2017, BioFactors.

[17]  S. Girardin,et al.  NLRX1 dampens oxidative stress and apoptosis in tissue injury via control of mitochondrial activity , 2017, The Journal of experimental medicine.

[18]  M. Laakso,et al.  Acute detachment of hexokinase II from mitochondria modestly increases oxygen consumption of the intact mouse heart. , 2017, Metabolism: clinical and experimental.

[19]  J. Bassaganya-Riera,et al.  NLRX1 Regulates Effector and Metabolic Functions of CD4+ T Cells , 2017, The Journal of Immunology.

[20]  A. Baartscheer,et al.  Reducing mitochondrial bound hexokinase II mediates transition from non-injurious into injurious ischemia/reperfusion of the intact heart , 2016, Journal of Physiology and Biochemistry.

[21]  Shanshan Zhang,et al.  NLRX1 attenuates apoptosis and inflammatory responses in myocardial ischemia by inhibiting MAVS-dependent NLRP3 inflammasome activation. , 2016, Molecular immunology.

[22]  C. Streutker,et al.  NLRX1 Acts as an Epithelial-Intrinsic Tumor Suppressor through the Modulation of TNF-Mediated Proliferation. , 2016, Cell reports.

[23]  G. Cline,et al.  Integrated, Step-Wise, Mass-Isotopomeric Flux Analysis of the TCA Cycle. , 2015, Cell metabolism.

[24]  G. Heusch Molecular basis of cardioprotection: signal transduction in ischemic pre-, post-, and remote conditioning. , 2015, Circulation research.

[25]  R. McPherson,et al.  Acylcarnitines: potential implications for skeletal muscle insulin resistance , 2015, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[26]  T. Hakvoort,et al.  Cardioprotective efficacy depends critically on pharmacological dose, duration of ischaemia, health status of animals and choice of anaesthetic regimen: a case study with folic acid , 2014, Journal of Translational Medicine.

[27]  M. Schultz,et al.  Nlrp3 plays no role in acute cardiac infarction due to low cardiac expression. , 2014, International journal of cardiology.

[28]  R. Southworth,et al.  Targeting hexokinase II to mitochondria to modulate energy metabolism and reduce ischaemia‐reperfusion injury in heart , 2014, British journal of pharmacology.

[29]  E. Liepinsh,et al.  The heart is better protected against myocardial infarction in the fed state compared to the fasted state. , 2014, Metabolism: clinical and experimental.

[30]  D. Philpott,et al.  NLRX1 does not inhibit MAVS-dependent antiviral signalling , 2013, Innate immunity.

[31]  J. Inserte,et al.  The role of mitochondrial permeability transition in reperfusion-induced cardiomyocyte death depends on the duration of ischemia , 2011, Basic Research in Cardiology.

[32]  P. Ranjan,et al.  NLRX1 protein attenuates inflammatory responses to infection by interfering with the RIG-I-MAVS and TRAF6-NF-κB signaling pathways. , 2011, Immunity.

[33]  M. Laakso,et al.  Disruption of Hexokinase II–Mitochondrial Binding Blocks Ischemic Preconditioning and Causes Rapid Cardiac Necrosis , 2011, Circulation research.

[34]  S. Nadtochiy,et al.  Cardioprotection by metabolic shut-down and gradual wake-up. , 2009, Journal of molecular and cellular cardiology.

[35]  T. Miura,et al.  Limitation of myocardial infarct size in the clinical setting: current status and challenges in translating animal experiments into clinical therapy , 2008, Basic research in cardiology.

[36]  M. Heise,et al.  NLRX1 is a regulator of mitochondrial antiviral immunity , 2008, Nature.

[37]  A. Meijer,et al.  Ischemic preconditioning, insulin, and morphine all cause hexokinase redistribution. , 2005, American journal of physiology. Heart and circulatory physiology.

[38]  M. Mocanu,et al.  Ischemic preconditioning protects by activating prosurvival kinases at reperfusion. , 2005, American journal of physiology. Heart and circulatory physiology.

[39]  S. Akira,et al.  Genetic depletion of cardiac myocyte STAT-3 abolishes classical preconditioning. , 2004, Cardiovascular research.

[40]  C. Des Rosiers,et al.  Profiling substrate fluxes in the isolated working mouse heart using 13C-labeled substrates: focusing on the origin and fate of pyruvate and citrate carbons. , 2004, American journal of physiology. Heart and circulatory physiology.

[41]  K. Shimamoto,et al.  Ischemic preconditioning activates AMPK in a PKC-dependent manner and induces GLUT4 up-regulation in the late phase of cardioprotection. , 2004, Cardiovascular research.

[42]  A. Leventhal,et al.  A national survey as a basis of public health policy: a case study with folic acid. , 2001, Public health reviews.

[43]  J. Mccormack,et al.  Regulation of myocardial carbohydrate metabolism under normal and ischaemic conditions Potential for pharmacological interventions , 1997 .