1
|
Michel LY, Esfahani H, De Mulder D, Verdoy R, Ambroise J, Roelants V, Bouchard B, Fabian N, Savary J, Dewulf JP, Doumont T, Bouzin C, Haufroid V, Luiken JJ, Nabben M, Singleton ML, Bertrand L, Ruiz M, Des Rosiers C, Balligand JL. An NRF2/β3-Adrenoreceptor Axis Drives a Sustained Antioxidant and Metabolic Rewiring Through the Pentose-Phosphate Pathway to Alleviate Cardiac Stress. Circulation 2025; 151:1312-1328. [PMID: 40071326 PMCID: PMC12052078 DOI: 10.1161/circulationaha.124.067876] [Citation(s) in RCA: 1] [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: 05/01/2024] [Accepted: 01/13/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Cardiac β3-adrenergic receptors (ARs) are upregulated in diseased hearts and mediate antithetic effects to those of β1AR and β2AR. β3AR agonists were recently shown to protect against myocardial remodeling in preclinical studies and to improve systolic function in patients with severe heart failure. However, the underlying mechanisms remain elusive. METHODS To dissect functional, transcriptional, and metabolic effects, hearts and isolated ventricular myocytes from mice harboring a moderate, cardiac-specific expression of a human ADRB3 transgene (β3AR-Tg) and subjected to transverse aortic constriction were assessed with echocardiography, RNA sequencing, positron emission tomography scan, metabolomics, and metabolic flux analysis. Subsequently, signaling and metabolic pathways were further investigated in vivo in β3AR-Tg and ex vivo in neonatal rat ventricular myocytes adenovirally infected to express β3AR and subjected to neurohormonal stress. These results were complemented with an analysis of single-nucleus RNA-sequencing data from human cardiac myocytes from patients with heart failure. RESULTS Compared with wild-type littermates, β3AR-Tg mice were protected from hypertrophy after transaortic constriction, and systolic function was preserved. β3AR-expressing hearts displayed enhanced myocardial glucose uptake under stress in the absence of increased lactate levels. Instead, metabolomic and metabolic flux analyses in stressed hearts revealed an increase in intermediates of the pentose-phosphate pathway in β3AR-Tg, an alternative route of glucose utilization, paralleled with increased transcript levels of NADPH-producing and rate-limiting enzymes of the pentose-phosphate pathway, without fueling the hexosamine metabolism. The ensuing increased content of NADPH and of reduced glutathione decreased myocyte oxidant stress, whereas downstream oxidative metabolism assessed by oxygen consumption was preserved with higher glucose oxidation in β3AR-Tg mice after transaortic constriction compared with wild type, together with increased mitochondrial biogenesis. Unbiased transcriptomics and pathway analysis identified NRF2 (NFE2L2) as an upstream transcription factor that was functionally verified in vivo and in β3AR-expressing cardiac myocytes, where its translocation and nuclear activity were dependent on β3AR activation of nitric oxide synthase and nitric oxide production through S-nitrosation of the NRF2-negative regulator Keap1. CONCLUSIONS Moderate expression of cardiac β3AR, at levels observed in human cardiac myocardium, exerts metabolic and antioxidant effects through activation of the pentose-phosphate pathway and NRF2 pathway through S-nitrosation of Keap1, thereby preserving myocardial oxidative metabolism, function, and integrity under pathophysiological stress.
Collapse
Affiliation(s)
- Lauriane Y.M. Michel
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St. Luc and Université catholique de Louvain, Brussels, Belgium (L.Y.M.M., H.E., D.d.M., R.V., N.F., J.-L.B.)
| | - Hrag Esfahani
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St. Luc and Université catholique de Louvain, Brussels, Belgium (L.Y.M.M., H.E., D.d.M., R.V., N.F., J.-L.B.)
| | - Delphine De Mulder
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St. Luc and Université catholique de Louvain, Brussels, Belgium (L.Y.M.M., H.E., D.d.M., R.V., N.F., J.-L.B.)
| | - Roxane Verdoy
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St. Luc and Université catholique de Louvain, Brussels, Belgium (L.Y.M.M., H.E., D.d.M., R.V., N.F., J.-L.B.)
| | - Jérôme Ambroise
- Institute of Experimental and Clinical Research (IREC), Centre des Technologies Moléculaires Appliquées, Université catholique de Louvain, Brussels, Belgium (J.A.)
| | - Véronique Roelants
- Institute of Experimental and Clinical Research (IREC), Molecular imagery, radiotherapy, oncology (MIRO), Cliniques Universitaires St. Luc and Université catholique de Louvain, Brussels, Belgium (V.R., T.D.)
- Nuclear Medicine Department, Cliniques Universitaires St. Luc and Université Catholique de Louvain, Brussels, Belgium (V.R.)
| | - Bertrand Bouchard
- Montreal Heart Institute Research Center, Quebec, Canada (B.B., M.R., C.D.R.)
| | - Nathalie Fabian
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St. Luc and Université catholique de Louvain, Brussels, Belgium (L.Y.M.M., H.E., D.d.M., R.V., N.F., J.-L.B.)
| | - Jérôme Savary
- Institute of Condensed Matter and Nanosciences (IMCN), Université catholique de Louvain, Louvain-la-Neuve, Belgium (J.S., M.L.S.)
| | - Joseph P. Dewulf
- Clinical Chemistry Department, Cliniques universitaires St. Luc and Louvain Centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Brussels, Belgium (J.P.D., V.H.)
| | - Thomas Doumont
- Institute of Experimental and Clinical Research (IREC), Molecular imagery, radiotherapy, oncology (MIRO), Cliniques Universitaires St. Luc and Université catholique de Louvain, Brussels, Belgium (V.R., T.D.)
| | - Caroline Bouzin
- 2IP-IREC Imaging Platform, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, RRID:SCR_023378, Brussels, Belgium (C.B.)
| | - Vincent Haufroid
- Clinical Chemistry Department, Cliniques universitaires St. Luc and Louvain Centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Brussels, Belgium (J.P.D., V.H.)
| | - Joost J.F.P. Luiken
- Departments of Genetics & Cell Biology and Clinical Genetics, Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center, Maastricht, the Netherlands (J.J.F.P.L., M.N.)
| | - Miranda Nabben
- Departments of Genetics & Cell Biology and Clinical Genetics, Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center, Maastricht, the Netherlands (J.J.F.P.L., M.N.)
| | - Michael L. Singleton
- Institute of Condensed Matter and Nanosciences (IMCN), Université catholique de Louvain, Louvain-la-Neuve, Belgium (J.S., M.L.S.)
| | - Luc Bertrand
- Institute of Experimental and Clinical Research (IREC), Pole of Cardiovascular Research (CARD), Université catholique de Louvain, Brussels, Belgium (L.B.)
- WELBIO Department, WEL Research Institute, Wavre, Belgium (L.B., J.-L.B.)
| | - Matthieu Ruiz
- Montreal Heart Institute Research Center, Quebec, Canada (B.B., M.R., C.D.R.)
- Department of Nutrition, Université de Montréal, Quebec, Canada (M.R., C.D.R.)
| | - Christine Des Rosiers
- Montreal Heart Institute Research Center, Quebec, Canada (B.B., M.R., C.D.R.)
- Department of Nutrition, Université de Montréal, Quebec, Canada (M.R., C.D.R.)
| | - Jean-Luc Balligand
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St. Luc and Université catholique de Louvain, Brussels, Belgium (L.Y.M.M., H.E., D.d.M., R.V., N.F., J.-L.B.)
- WELBIO Department, WEL Research Institute, Wavre, Belgium (L.B., J.-L.B.)
| |
Collapse
|
2
|
Tatekoshi Y, Mahmoodzadeh A, Shapiro JS, Liu M, Bianco GM, Tatekoshi A, Camp SD, De Jesus A, Koleini N, De La Torre S, Wasserstrom JA, Dillmann WH, Thomson BR, Bedi KC, Margulies KB, Weinberg SE, Ardehali H. Protein O-GlcNAcylation and hexokinase mitochondrial dissociation drive heart failure with preserved ejection fraction. Cell Metab 2025:S1550-4131(25)00211-6. [PMID: 40267914 DOI: 10.1016/j.cmet.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 03/03/2025] [Accepted: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a common cause of morbidity and mortality worldwide, but its pathophysiology remains unclear. Here, we report a mouse model of HFpEF and show that hexokinase (HK)-1 mitochondrial binding in endothelial cells (ECs) is critical for protein O-GlcNAcylation and the development of HFpEF. We demonstrate increased mitochondrial dislocation of HK1 within ECs in HFpEF mice. Mice with deletion of the mitochondrial-binding domain of HK1 spontaneously develop HFpEF and display impaired angiogenesis. Spatial proximity of dislocated HK1 and O-linked N-acetylglucosamine transferase (OGT) causes increased OGT activity, shifting the balance of the hexosamine biosynthetic pathway intermediates into the O-GlcNAcylation machinery. EC-specific overexpression of O-GlcNAcase and an OGT inhibitor reverse angiogenic defects and the HFpEF phenotype, highlighting the importance of protein O-GlcNAcylation in the development of HFpEF. Our study demonstrates a new mechanism for HFpEF through HK1 cellular localization and resultant protein O-GlcNAcylation, and provides a potential therapy for HFpEF.
Collapse
Affiliation(s)
- Yuki Tatekoshi
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Amir Mahmoodzadeh
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Jason S Shapiro
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Mingyang Liu
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - George M Bianco
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Ayumi Tatekoshi
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Spencer Duncan Camp
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Adam De Jesus
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Navid Koleini
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Santiago De La Torre
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - J Andrew Wasserstrom
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA
| | - Wolfgang H Dillmann
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Benjamin R Thomson
- Feinberg Cardiovascular and Renal Research Institute and Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kenneth C Bedi
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Samuel E Weinberg
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hossein Ardehali
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
3
|
Liffers JL, Reinhardt JP, Seidl MD, Kirchhefer U, Müller FU, Schulte JS. Cre recombinase affects calcium dynamics already in young mice. Front Pharmacol 2025; 16:1558573. [PMID: 40206076 PMCID: PMC11979376 DOI: 10.3389/fphar.2025.1558573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
Background The Cre/LoxP system is widely used in cardiovascular research to generate mouse models with tissue-specific inactivation of target genes. Studies have reported that expression of Cre recombinase under the αMHC promoter leads to age-dependent cardiotoxicity with ventricular hypertrophy, fibrosis and ventricular dysfunction at 6 months of age. This study explores the impact of Cre expression on intracellular Ca2+ dynamics in ventricular myocytes of αMHC-Cre mice as early as 3 months old. Methods Mice expressing Cre under the αMHC promoter (CRE) were compared to wild-type (WT) controls. Ventricular cardiomyocytes (VCMs) were isolated by the Langendorff method. Ca2+ transients and sarcomere shortening were simultaneously recorded from VCMs. Ventricular and atrial weights were assessed, VCM dimensions analyzed, and protein and mRNA levels of key proteins involved in Ca2+ dynamics measured by immunoblot analysis and quantitative real-time RT-PCR. Results At 3 months, CRE mice showed no evidence of cardiac hypertrophy. Ventricular or atrial weights and VCM size were not different between CRE and WT mice. The same applied to protein levels of SERCA2a, NCX1, Cav1.2, PLN and its phosphorylated form PLN pThr17. Nevertheless Ca2+ dynamics were significantly altered in CRE mice. Under basal conditions resting and peak Ca2+ were reduced and Ca2+ transient decay was delayed up to 30% in VCMs from CRE vs. WT mice. These differences persisted upon stimulation with 1 µM isoproterenol, whereas Ca2+ transient amplitude increased in CRE VCMs. We confirmed a previously reported reduction in dystrophin, potentially explaining the changes in Ca2+ dynamics. Despite these changes sarcomere shortening parameters were not different between groups. Conclusion As early as 3 months of age, Cre expression in VCMs leads to changes in Ca2+ dynamics that do not yet affect sarcomere shortening and cannot be attributed to the regulation of key proteins involved in Ca2+ dynamics. Because changes in intracellular Ca2+ dynamics can affect gene expression through altered excitation-transcription coupling, researchers should be aware of these subtle changes that precede the prominent phenotype at 6 months of age. Therefore, it is essential to use Cre-positive mice as controls when analyzing knockout models generated by the Cre/LoxP system.
Collapse
|
4
|
Keller MA, Nakamura M. Ketone Catabolism is Essential for Maintaining Normal Heart Function During Aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643760. [PMID: 40161834 PMCID: PMC11952331 DOI: 10.1101/2025.03.18.643760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The heart utilizes various nutrient sources for energy production, primarily favoring fatty acid oxidation. While ketones can be fuel substrates, ketolysis has been shown to be dispensable for heart development and function in mice. However, the long-term consequences of ketolysis downregulation in the heart remain unknown. Here we demonstrate that ketone catabolism is essential for preserving cardiac function during aging. The cardiac expression of succinyl-CoA:3-ketoacid CoA transferase (SCOT), a rate-limiting enzyme in ketolysis, decreases with aging in female mice. SCOT cardiomyocyte-specific knockout (cKO) mice exhibit normal heart function at 10 weeks of age but progressively develop cardiac dysfunction and remodeling as they age, without overt hypertrophy in both sexes. Notably, ketone supplementation via a ketogenic diet partially rescues contractile dysfunction in SCOT cKO mice, suggesting ketone oxidation-independent mechanisms contribute to the development of cardiomyopathy caused by SCOT downregulation. These findings indicate that ketone catabolism is crucial for maintaining heart function during aging, and that ketones confer cardioprotection independently of ketone oxidation.
Collapse
|
5
|
Weeks KL, McMullen JR. Cardiomyocyte-Specific FoxO1 Knockout Mice as Tools to Assess Cardiac Hypertrophy and Key Experimental Considerations Using Cre-loxP. Methods Mol Biol 2025; 2871:69-81. [PMID: 39565579 DOI: 10.1007/978-1-0716-4217-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
The generation of tissue-specific mouse models has provided a powerful strategy to understand the role of genes in specific tissues/cells of interest under control/basal conditions and in response to physiological and pathological stimuli. Here we describe the generation of cardiomyocyte-specific FoxO1 knockout mice using Cre-loxP technology to examine the role of FoxO1 for the induction of heart enlargement (cardiac hypertrophy) in settings of health and disease. We highlight breeding strategies for generating tissue-specific mouse models and key experimental considerations during characterization.
Collapse
Affiliation(s)
- Kate L Weeks
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia.
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC, Australia.
- Central Clinical School, Monash University, Clayton, VIC, Australia.
| | - Julie R McMullen
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC, Australia.
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia.
- Monash Alfred Baker Centre for Cardiovascular Research, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.
- Heart Research Institute, Sydney, N.S.W., Australia.
| |
Collapse
|
6
|
Sermersheim TJ, Phillips LJ, Evans PL, Kahn BB, Welc SS, Witczak CA. Regulation of injury-induced skeletal myofiber regeneration by glucose transporter 4 (GLUT4). Skelet Muscle 2024; 14:33. [PMID: 39695900 DOI: 10.1186/s13395-024-00366-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Insulin resistance and type 2 diabetes impair cellular regeneration in multiple tissues including skeletal muscle. The molecular basis for this impairment is largely unknown. Glucose uptake via glucose transporter GLUT4 is impaired in insulin resistance. In healthy muscle, acute injury stimulates glucose uptake. Whether decreased glucose uptake via GLUT4 impairs muscle regeneration is presently unknown. The goal of this study was to determine whether GLUT4 regulates muscle glucose uptake and/or regeneration following acute injury. METHODS Tibialis anterior and extensor digitorum longus muscles from wild-type, control, or muscle-specific GLUT4 knockout (mG4KO) mice were injected with the myotoxin barium chloride to induce muscle injury. After 3, 5, 7, 10, 14, or 21 days (in wild-type mice), or after 7 or 14 days (in control & mG4KO) mice, muscles were isolated to examine [3H]-2-deoxyglucose uptake, GLUT4 levels, extracellular fluid space, fibrosis, myofiber cross-sectional area, and myofiber centralized nuclei. RESULTS In wild-type mice, muscle glucose uptake was increased 3, 5, 7, and 10 days post-injury. There was a rapid decrease in GLUT4 protein levels that were restored to baseline at 5-7 days post-injury, followed by a super-compensation at 10-21 days. In mG4KO mice, there were no differences in muscle glucose uptake, extracellular fluid space, muscle fibrosis, myofiber cross-sectional areas, or percentage of centrally nucleated myofibers at 7 days post-injury. In contrast, at 14 days injured muscles from mG4KO mice exhibited decreased glucose uptake, muscle weight, myofiber cross sectional areas, and centrally nucleated myofibers, with no change in extracellular fluid space or fibrosis. CONCLUSIONS Collectively, these findings demonstrate that glucose uptake via GLUT4 regulates skeletal myofiber regeneration following acute injury.
Collapse
Affiliation(s)
- Tyler J Sermersheim
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Diabetes & Metabolic Diseases, Indianapolis, IN, USA
| | - LeAnna J Phillips
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Diabetes & Metabolic Diseases, Indianapolis, IN, USA
| | - Parker L Evans
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Diabetes & Metabolic Diseases, Indianapolis, IN, USA
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes & Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Steven S Welc
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA
| | - Carol A Witczak
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Center for Diabetes & Metabolic Diseases, Indianapolis, IN, USA.
| |
Collapse
|
7
|
Shi Q, Wang J, Malik H, Li X, Streeter J, Sharafuddin J, Weatherford E, Stein D, Itan Y, Chen B, Hall D, Song LS, Abel ED. IRS2 Signaling Protects Against Stress-Induced Arrhythmia by Maintaining Ca 2+ Homeostasis. Circulation 2024; 150:1966-1983. [PMID: 39253856 PMCID: PMC11631690 DOI: 10.1161/circulationaha.123.065048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/13/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND The docking protein IRS2 (insulin receptor substrate protein-2) is an important mediator of insulin signaling and may also regulate other signaling pathways. Murine hearts with cardiomyocyte-restricted deletion of Irs2 (cIRS2-KO) are more susceptible to pressure overload-induced cardiac dysfunction, implying a critical protective role of IRS2 in cardiac adaptation to stress through mechanisms that are not fully understood. There is limited evidence regarding the function of IRS2 beyond metabolic homeostasis regulation, particularly in the context of cardiac disease. METHODS A retrospective analysis of an electronic medical record database was conducted to identify patients with IRS2 variants and assess their risk of cardiac arrhythmias. Arrhythmia susceptibility was examined in cIRS2-KO mice. The underlying mechanisms were investigated using confocal calcium imaging of ex vivo whole hearts and isolated cardiomyocytes to assess calcium handling, Western blotting to analyze the involved signaling pathways, and pharmacological and genetic interventions to rescue arrhythmias in cIRS2-KO mice. RESULTS The retrospective analysis identified patients with IRS2 variants of uncertain significance with a potential association to an increased risk of cardiac arrhythmias compared with matched controls. cIRS2-KO hearts were found to be prone to catecholamine-sensitive ventricular tachycardia and reperfusion ventricular tachycardia. Confocal calcium imaging of ex vivo whole hearts and single isolated cardiomyocytes from cIRS2-KO hearts revealed decreased Ca²⁺ transient amplitudes, increased spontaneous Ca²⁺ sparks, and reduced sarcoplasmic reticulum Ca²⁺ content during sympathetic stress, indicating sarcoplasmic reticulum dysfunction. We identified that overactivation of the AKT1/NOS3 (nitric oxide synthase 3)/CaMKII (Ca²⁺/calmodulin-dependent protein kinase II)/RyR2 (type 2 ryanodine receptor) signaling pathway led to calcium mishandling and catecholamine-sensitive ventricular tachycardia in cIRS2-KO hearts. Pharmacological AKT inhibition or genetic stabilization of RyR2 rescued catecholamine-sensitive ventricular tachycardia in cIRS2-KO mice. CONCLUSIONS Cardiac IRS2 inhibits sympathetic stress-induced AKT/NOS3/CaMKII/RyR2 overactivation and calcium-dependent arrhythmogenesis. This novel IRS2 signaling axis, essential for maintaining cardiac calcium homeostasis under stress, presents a promising target for developing new antiarrhythmic therapies.
Collapse
Affiliation(s)
- Qian Shi
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Jinxi Wang
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Hamza Malik
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Xuguang Li
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Jennifer Streeter
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Jacob Sharafuddin
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Eric Weatherford
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - David Stein
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Genetics and Genome Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yuval Itan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Genetics and Genome Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Biyi Chen
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Duane Hall
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Long-Sheng Song
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - E. Dale Abel
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
- Current address, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA
| |
Collapse
|
8
|
Harrison AG, Yang D, Cahoon JG, Geng T, Cao Z, Karginov TA, Hu Y, Li X, Chiari CC, Qyang Y, Vella AT, Fan Z, Vanaja SK, Rathinam VA, Witczak CA, Bogan JS, Wang P. UBXN9 governs GLUT4-mediated spatial confinement of RIG-I-like receptors and signaling. Nat Immunol 2024; 25:2234-2246. [PMID: 39567760 PMCID: PMC12067455 DOI: 10.1038/s41590-024-02004-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/04/2024] [Indexed: 11/22/2024]
Abstract
The cytoplasmic RIG-I-like receptors (RLRs) recognize viral RNA and initiate innate antiviral immunity. RLR signaling also triggers glycolytic reprogramming through glucose transporters (GLUTs), whose role in antiviral immunity is elusive. Here, we unveil that insulin-responsive GLUT4 inhibits RLR signaling independently of glucose uptake in adipose and muscle tissues. At steady state, GLUT4 is trapped at the Golgi matrix by ubiquitin regulatory X domain 9 (UBXN9, TUG). Following RNA virus infection, GLUT4 is released and translocated to the cell surface where it spatially segregates a significant pool of cytosolic RLRs, preventing them from activating IFN-β responses. UBXN9 deletion prompts constitutive GLUT4 translocation, sequestration of RLRs and attenuation of antiviral immunity, whereas GLUT4 deletion heightens RLR signaling. Notably, reduced GLUT4 expression is uniquely associated with human inflammatory myopathies characterized by hyperactive interferon responses. Overall, our results demonstrate a noncanonical UBXN9-GLUT4 axis that controls antiviral immunity via plasma membrane tethering of cytosolic RLRs.
Collapse
Affiliation(s)
- Andrew G Harrison
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Duomeng Yang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA.
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, Guangdong, China.
| | - Jason G Cahoon
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Tingting Geng
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Ziming Cao
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Timofey A Karginov
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Youjia Hu
- Section of Endocrinology, Department of Internal Medicine, and Department of Cell Biology, and Yale Center for Molecular and Systems Metabolism, Yale School of Medicine, New Haven, CT, USA
| | - Xin Li
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Conner C Chiari
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Yibing Qyang
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Anthony T Vella
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Zhichao Fan
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Sivapriya Kailasan Vanaja
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Vijay A Rathinam
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Carol A Witczak
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jonathan S Bogan
- Section of Endocrinology, Department of Internal Medicine, and Department of Cell Biology, and Yale Center for Molecular and Systems Metabolism, Yale School of Medicine, New Haven, CT, USA
| | - Penghua Wang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
9
|
Su S, Quan C, Chen Q, Wang R, Du Q, Zhu S, Li M, Yang X, Rong P, Chen J, Bai Y, Zheng W, Feng W, Liu M, Xie B, Ouyang K, Shi YS, Lan F, Zhang X, Xiao R, Chen X, Wang HY, Chen S. AS160 is a lipid-responsive regulator of cardiac Ca 2+ homeostasis by controlling lysophosphatidylinositol metabolism and signaling. Nat Commun 2024; 15:9602. [PMID: 39505896 PMCID: PMC11542008 DOI: 10.1038/s41467-024-54031-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
The obese heart undergoes metabolic remodeling and exhibits impaired calcium (Ca2+) homeostasis, which are two critical assaults leading to cardiac dysfunction. The molecular mechanisms underlying these alterations in obese heart are not well understood. Here, we show that the Rab-GTPase activating protein AS160 is a lipid-responsive regulator of Ca2+ homeostasis through governing lysophosphatidylinositol metabolism and signaling. Palmitic acid/high fat diet inhibits AS160 activity through phosphorylation by NEK6, which consequently activates its downstream target Rab8a. Inactivation of AS160 in cardiomyocytes elevates cytosolic Ca2+ that subsequently impairs cardiac contractility. Mechanistically, Rab8a downstream of AS160 interacts with DDHD1 to increase lysophosphatidylinositol metabolism and signaling that leads to Ca2+ release from sarcoplasmic reticulum. Inactivation of NEK6 prevents inhibition of AS160 by palmitic acid/high fat diet, and alleviates cardiac dysfunction in high fat diet-fed mice. Together, our findings reveal a regulatory mechanism governing metabolic remodeling and Ca2+ homeostasis in obese heart, and have therapeutic implications to combat obesity cardiomyopathy.
Collapse
Affiliation(s)
- Shu Su
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Chao Quan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Qiaoli Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Ruizhen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Qian Du
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Sangsang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Min Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Xinyu Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Ping Rong
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Jiang Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yingyu Bai
- Department of Biopharmaceuticals & Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wen Zheng
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Weikuan Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Minjun Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Bingxian Xie
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yun Stone Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Feng Lan
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiuqin Zhang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Ruiping Xiao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Xiongwen Chen
- Department of Biopharmaceuticals & Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Hong-Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
- Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China.
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Nanjing, China.
| |
Collapse
|
10
|
Davenport A, Kessinger CW, Pfeiffer RD, Shah N, Xu R, Abel ED, Tucker NR, Lin Z. Comparative analysis of two independent Myh6-Cre transgenic mouse lines. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 9:100081. [PMID: 39323506 PMCID: PMC11423776 DOI: 10.1016/j.jmccpl.2024.100081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 09/27/2024]
Abstract
We have previously shown that the Myh6 promoter drives Cre expression in a subset of male germ line cells in three independent Myh6-Cre mouse lines, including two transgenic lines and one knock-in allele. In this study, we further compared the tissue-specificity of the two Myh6-Cre transgenic mouse lines, MDS Myh6-Cre and AUTR Myh6-Cre, through examining the expression of tdTomato (tdTom) red fluorescence protein in multiple internal organs, including the heart, brain, liver, lung, pancreas and brown adipose tissue. Our results show that MDS Myh6-Cre mainly activates tdTom reporter in the heart, whereas AUTR Myh6-Cre activates tdTom expression significantly in the heart, and in the cells of liver, pancreas and brain. In the heart, similar to MDS Myh6-Cre, AUTR Myh6-Cre activates tdTom in most cardiomyocytes. In the other organs, AUTR Myh6-Cre not only mosaically activates tdTom in some parenchymal cells, such as hepatocytes in the liver and neurons in the brain, but also turns on tdTom in some interstitial cells of unknown identity.
Collapse
Affiliation(s)
- Amanda Davenport
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - Chase W. Kessinger
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - Ryan D. Pfeiffer
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - Nikita Shah
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
- College of Arts and Sciences, SUNY Polytechnic Institute, Utica, NY 13502, United States of America
| | - Richard Xu
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - E. Dale Abel
- Department of Medicine David Geffen School of Medicine and UCLA Health, United States of America
| | - Nathan R. Tucker
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - Zhiqiang Lin
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| |
Collapse
|
11
|
Gural B, Kirkland L, Hockett A, Sandroni P, Zhang J, Rosa-Garrido M, Swift SK, Chapski D, Flinn MA, O'Meara CC, Vondriska TM, Patterson M, Jensen BC, Rau CD. Novel Insights into Post-Myocardial Infarction Cardiac Remodeling through Algorithmic Detection of Cell-Type Composition Shifts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607400. [PMID: 39149394 PMCID: PMC11326268 DOI: 10.1101/2024.08.09.607400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Background Recent advances in single cell sequencing have led to an increased focus on the role of cell-type composition in phenotypic presentation and disease progression. Cell-type composition research in the heart is challenging due to large, frequently multinucleated cardiomyocytes that preclude most single cell approaches from obtaining accurate measurements of cell composition. Our in silico studies reveal that ignoring cell type composition when calculating differentially expressed genes (DEGs) can have significant consequences. For example, a relatively small change in cell abundance of only 10% can result in over 25% of DEGs being false positives. Methods We have implemented an algorithmic approach that uses snRNAseq datasets as a reference to accurately calculate cell type compositions from bulk RNAseq datasets through robust data cleaning, gene selection, and multi-sample cross-subject and cross-cell-type deconvolution. We applied our approach to cardiomyocyte-specific α1A adrenergic receptor (CM-α1A-AR) knockout mice. 8-12 week-old mice (either WT or CM-α1A-KO) were subjected to permanent left coronary artery (LCA) ligation or sham surgery (n=4 per group). Transcriptomes from the infarct border zones were collected 3 days later and analyzed using our algorithm to determine cell-type abundances, corrected differential expression calculations using DESeq2, and validated these findings using RNAscope. Results Uncorrected DEGs for the CM-α1A-KO X LCA interaction term featured many cell-type specific genes such as Timp4 (fibroblasts) and Aplnr (cardiomyocytes) and overall GO enrichment for terms pertaining to cardiomyocyte differentiation (P=3.1E-4). Using our algorithm, we observe a striking loss of cardiomyocytes and gain in fibroblasts in the α1A-KO + LCA mice that was not recapitulated in WT + LCA animals, although we did observe a similar increase in macrophage abundance in both conditions. This recapitulates prior results that showed a much more severe heart failure phenotype in CM-α1A-KO + LCA mice. Following correction for cell-type, our DEGs now highlight a novel set of genes enriched for GO terms such as cardiac contraction (P=3.7E-5) and actin filament organization (P=6.3E-5). Conclusions Our algorithm identifies and corrects for cell-type abundance in bulk RNAseq datasets opening new avenues for research on novel genes and pathways as well as an improved understanding of the role of cardiac cell types in cardiovascular disease.
Collapse
Affiliation(s)
- Brian Gural
- Department of Genetics and Computational Medicine Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Logan Kirkland
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, Division of Cardiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Abbey Hockett
- Department of Genetics and Computational Medicine Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peyton Sandroni
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiandong Zhang
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, Division of Cardiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Manuel Rosa-Garrido
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Samantha K Swift
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Douglas Chapski
- Departments of Anesthesiology & Perioperative Medicine, Medicine/Cardiology, and Physiology, David Geffen School of Medicine; Molecular Biology Institute; University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael A Flinn
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Caitlin C O'Meara
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Thomas M Vondriska
- Departments of Anesthesiology & Perioperative Medicine, Medicine/Cardiology, and Physiology, David Geffen School of Medicine; Molecular Biology Institute; University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michaela Patterson
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Brian C Jensen
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, Division of Cardiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Christoph D Rau
- Department of Genetics and Computational Medicine Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Alibhai FJ, Li RK. Rejuvenation of the Aging Heart: Molecular Determinants and Applications. Can J Cardiol 2024; 40:1394-1411. [PMID: 38460612 DOI: 10.1016/j.cjca.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024] Open
Abstract
In Canada and worldwide, the elderly population (ie, individuals > 65 years of age) is increasing disproportionately relative to the total population. This is expected to have a substantial impact on the health care system, as increased aged is associated with a greater incidence of chronic noncommunicable diseases. Within the elderly population, cardiovascular disease is a leading cause of death, therefore developing therapies that can prevent or slow disease progression in this group is highly desirable. Historically, aging research has focused on the development of anti-aging therapies that are implemented early in life and slow the age-dependent decline in cell and organ function. However, accumulating evidence supports that late-in-life therapies can also benefit the aged cardiovascular system by limiting age-dependent functional decline. Moreover, recent studies have demonstrated that rejuvenation (ie, reverting cellular function to that of a younger phenotype) of the already aged cardiovascular system is possible, opening new avenues to develop therapies for older individuals. In this review, we first provide an overview of the functional changes that occur in the cardiomyocyte with aging and how this contributes to the age-dependent decline in heart function. We then discuss the various anti-aging and rejuvenation strategies that have been pursued to improve the function of the aged cardiomyocyte, with a focus on therapies implemented late in life. These strategies include 1) established systemic approaches (caloric restriction, exercise), 2) pharmacologic approaches (mTOR, AMPK, SIRT1, and autophagy-targeting molecules), and 3) emerging rejuvenation approaches (partial reprogramming, parabiosis/modulation of circulating factors, targeting endogenous stem cell populations, and senotherapeutics). Collectively, these studies demonstrate the exciting potential and limitations of current rejuvenation strategies and highlight future areas of investigation that will contribute to the development of rejuvenation therapies for the aged heart.
Collapse
Affiliation(s)
- Faisal J Alibhai
- Toronto General Research Hospital Institute, University Health Network, Toronto, Ontario, Canada
| | - Ren-Ke Li
- Toronto General Research Hospital Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery, Division of Cardiovascular Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
13
|
Caturano A, Galiero R, Vetrano E, Sardu C, Rinaldi L, Russo V, Monda M, Marfella R, Sasso FC. Insulin-Heart Axis: Bridging Physiology to Insulin Resistance. Int J Mol Sci 2024; 25:8369. [PMID: 39125938 PMCID: PMC11313400 DOI: 10.3390/ijms25158369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Insulin signaling is vital for regulating cellular metabolism, growth, and survival pathways, particularly in tissues such as adipose, skeletal muscle, liver, and brain. Its role in the heart, however, is less well-explored. The heart, requiring significant ATP to fuel its contractile machinery, relies on insulin signaling to manage myocardial substrate supply and directly affect cardiac muscle metabolism. This review investigates the insulin-heart axis, focusing on insulin's multifaceted influence on cardiac function, from metabolic regulation to the development of physiological cardiac hypertrophy. A central theme of this review is the pathophysiology of insulin resistance and its profound implications for cardiac health. We discuss the intricate molecular mechanisms by which insulin signaling modulates glucose and fatty acid metabolism in cardiomyocytes, emphasizing its pivotal role in maintaining cardiac energy homeostasis. Insulin resistance disrupts these processes, leading to significant cardiac metabolic disturbances, autonomic dysfunction, subcellular signaling abnormalities, and activation of the renin-angiotensin-aldosterone system. These factors collectively contribute to the progression of diabetic cardiomyopathy and other cardiovascular diseases. Insulin resistance is linked to hypertrophy, fibrosis, diastolic dysfunction, and systolic heart failure, exacerbating the risk of coronary artery disease and heart failure. Understanding the insulin-heart axis is crucial for developing therapeutic strategies to mitigate the cardiovascular complications associated with insulin resistance and diabetes.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Luca Rinaldi
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy;
| | - Vincenzo Russo
- Department of Biology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA;
- Division of Cardiology, Department of Medical Translational Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| |
Collapse
|
14
|
Wang P, Harrison A, Yang D, Cahoon J, Geng T, Cao Z, Karginov T, Chiari C, Li X, Qyang Y, Vella A, Fan Z, Vanaja SK, Rathinam V, Witczak C, Bogan J. UBXN9 governs GLUT4-mediated spatial confinement of RIG-I-like receptors and signaling. RESEARCH SQUARE 2024:rs.3.rs-3373803. [PMID: 38883790 PMCID: PMC11177981 DOI: 10.21203/rs.3.rs-3373803/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The cytoplasmic RIG-I-like receptors (RLRs) recognize viral RNA and initiate innate antiviral immunity. RLR signaling also triggers glycolytic reprogramming through glucose transporters (GLUTs), whose role in antiviral immunity is elusive. Here, we unveil that insulin-responsive GLUT4 inhibits RLR signaling independently of glucose uptake in adipose and muscle tissues. At steady state, GLUT4 is docked at the Golgi matrix by ubiquitin regulatory X domain 9 (UBXN9, TUG). Following RNA virus infection, GLUT4 is released and translocated to the cell surface where it spatially segregates a significant pool of cytosolic RLRs, preventing them from activating IFN-β responses. UBXN9 deletion prompts constitutive GLUT4 trafficking, sequestration of RLRs, and attenuation of antiviral immunity, whereas GLUT4 deletion heightens RLR signaling. Notably, reduced GLUT4 expression is uniquely associated with human inflammatory myopathies characterized by hyperactive interferon responses. Overall, our results demonstrate a noncanonical UBXN9-GLUT4 axis that controls antiviral immunity via plasma membrane tethering of cytosolic RLRs.
Collapse
|
15
|
Shen T, Wang S, Huang C, Zhu S, Zhu X, Li N, Wang H, Huang L, Ren M, Han Z, Ge J, Chen Z, Ouyang K. Cardiac-specific deletion of heat shock protein 60 induces mitochondrial stress and disrupts heart development in mice. Biochem Biophys Res Commun 2024; 710:149883. [PMID: 38588611 DOI: 10.1016/j.bbrc.2024.149883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024]
Abstract
Congenital heart diseases are the most common birth defects around the world. Emerging evidence suggests that mitochondrial homeostasis is required for normal heart development. In mitochondria, a series of molecular chaperones including heat shock protein 60 (HSP60) are engaged in assisting the import and folding of mitochondrial proteins. However, it remains largely obscure whether and how these mitochondrial chaperones regulate cardiac development. Here, we generated a cardiac-specific Hspd1 deletion mouse model by αMHC-Cre and investigated the role of HSP60 in cardiac development. We observed that deletion of HSP60 in embryonic cardiomyocytes resulted in abnormal heart development and embryonic lethality, characterized by reduced cardiac cell proliferation and thinner ventricular walls, highlighting an essential role of cardiac HSP60 in embryonic heart development and survival. Our results also demonstrated that HSP60 deficiency caused significant downregulation of mitochondrial ETC subunits and induced mitochondrial stress. Analysis of gene expression revealed that P21 that negatively regulates cell proliferation is significantly upregulated in HSP60 knockout hearts. Moreover, HSP60 deficiency induced activation of eIF2α-ATF4 pathway, further indicating the underlying mitochondrial stress in cardiomyocytes after HSP60 deletion. Taken together, our study demonstrated that regular function of mitochondrial chaperones is pivotal for maintaining normal mitochondrial homeostasis and embryonic heart development.
Collapse
Affiliation(s)
- Tao Shen
- Department of Cardiovascular Surgery, Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui province, China; Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong province, China
| | - Shuting Wang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, Guangdong province, China
| | - Can Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong province, China
| | - Siting Zhu
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, Guangdong province, China; Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Xiangbin Zhu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong province, China
| | - Na Li
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong province, China
| | - Hong Wang
- Central Laboratory, Peking University Shenzhen Hospital, China
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong province, China
| | - Mingming Ren
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong province, China
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong province, China
| | - Jianjun Ge
- Department of Cardiovascular Surgery, Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui province, China.
| | - Ze'e Chen
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong province, China.
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong province, China.
| |
Collapse
|
16
|
Xia W, Zhang M, Liu C, Wang S, Xu A, Xia Z, Pang L, Cai Y. Exploring the therapeutic potential of tetrahydrobiopterin for heart failure with preserved ejection fraction: A path forward. Life Sci 2024; 345:122594. [PMID: 38537900 DOI: 10.1016/j.lfs.2024.122594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/10/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
A large number of patients are affected by classical heart failure (HF) symptomatology with preserved ejection fraction (HFpEF) and multiorgan syndrome. Due to high morbidity and mortality rate, hospitalization and mortality remain serious socioeconomic problems, while the lack of effective pharmacological or device treatment means that HFpEF presents a major unmet medical need. Evidence from clinical and basic studies demonstrates that systemic inflammation, increased oxidative stress, and impaired mitochondrial function are the common pathological mechanisms in HFpEF. Tetrahydrobiopterin (BH4), beyond being an endogenous co-factor for catalyzing the conversion of some essential biomolecules, has the capacity to prevent systemic inflammation, enhance antioxidant resistance, and modulate mitochondrial energy production. Therefore, BH4 has emerged in the last decade as a promising agent to prevent or reverse the progression of disorders such as cardiovascular disease. In this review, we cover the clinical progress and limitations of using downstream targets of nitric oxide (NO) through NO donors, soluble guanylate cyclase activators, phosphodiesterase inhibitors, and sodium-glucose co-transporter 2 inhibitors in treating cardiovascular diseases, including HFpEF. We discuss the use of BH4 in association with HFpEF, providing new evidence for its potential use as a pharmacological option for treating HFpEF.
Collapse
Affiliation(s)
- Weiyi Xia
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Miao Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Guangdong, China
| | - Chang Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Lei Pang
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China.
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
17
|
Shi Q, Malik H, Crawford RM, Streeter J, Wang J, Huo R, Shih JC, Chen B, Hall D, Abel ED, Song LS, Anderson EJ. Cardiac monoamine oxidase-A inhibition protects against catecholamine-induced ventricular arrhythmias via enhanced diastolic calcium control. Cardiovasc Res 2024; 120:596-611. [PMID: 38198753 PMCID: PMC11074799 DOI: 10.1093/cvr/cvae012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 11/01/2023] [Accepted: 11/22/2023] [Indexed: 01/12/2024] Open
Abstract
AIMS A mechanistic link between depression and risk of arrhythmias could be attributed to altered catecholamine metabolism in the heart. Monoamine oxidase-A (MAO-A), a key enzyme involved in catecholamine metabolism and longstanding antidepressant target, is highly expressed in the myocardium. The present study aimed to elucidate the functional significance and underlying mechanisms of cardiac MAO-A in arrhythmogenesis. METHODS AND RESULTS Analysis of the TriNetX database revealed that depressed patients treated with MAO inhibitors had a lower risk of arrhythmias compared with those treated with selective serotonin reuptake inhibitors. This effect was phenocopied in mice with cardiomyocyte-specific MAO-A deficiency (cMAO-Adef), which showed a significant reduction in both incidence and duration of catecholamine stress-induced ventricular tachycardia compared with wild-type mice. Additionally, cMAO-Adef cardiomyocytes exhibited altered Ca2+ handling under catecholamine stimulation, with increased diastolic Ca2+ reuptake, reduced diastolic Ca2+ leak, and diminished systolic Ca2+ release. Mechanistically, cMAO-Adef hearts had reduced catecholamine levels under sympathetic stress, along with reduced levels of reactive oxygen species and protein carbonylation, leading to decreased oxidation of Type II PKA and CaMKII. These changes potentiated phospholamban (PLB) phosphorylation, thereby enhancing diastolic Ca2+ reuptake, while reducing ryanodine receptor 2 (RyR2) phosphorylation to decrease diastolic Ca2+ leak. Consequently, cMAO-Adef hearts exhibited lower diastolic Ca2+ levels and fewer arrhythmogenic Ca2+ waves during sympathetic overstimulation. CONCLUSION Cardiac MAO-A inhibition exerts an anti-arrhythmic effect by enhancing diastolic Ca2+ handling under catecholamine stress.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Calcium/metabolism
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Catecholamines/metabolism
- Cells, Cultured
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Diastole/drug effects
- Disease Models, Animal
- Heart Rate/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Monoamine Oxidase/metabolism
- Monoamine Oxidase Inhibitors/pharmacology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phosphorylation
- Reactive Oxygen Species/metabolism
- Ryanodine Receptor Calcium Release Channel/metabolism
- Tachycardia, Ventricular/enzymology
- Tachycardia, Ventricular/physiopathology
Collapse
Affiliation(s)
- Qian Shi
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Hamza Malik
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Rachel M Crawford
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 180 S Grand Ave., Iowa City, IA 52242, USA
| | - Jennifer Streeter
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Jinxi Wang
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Ran Huo
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 180 S Grand Ave., Iowa City, IA 52242, USA
| | - Jean C Shih
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Biyi Chen
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Duane Hall
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
| | - E Dale Abel
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, 169 Newton Rd, Iowa City, IA 52242, USA
| | - Long-Sheng Song
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, 169 Newton Rd, Iowa City, IA 52242, USA
| | - Ethan J Anderson
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 180 S Grand Ave., Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, 169 Newton Rd, Iowa City, IA 52242, USA
| |
Collapse
|
18
|
Oliver T, Nguyen NY, Tully CB, McCormack NM, Sun CM, Fiorillo AA, Heier CR. The glucocorticoid receptor acts locally to protect dystrophic muscle and heart during disease. Dis Model Mech 2024; 17:dmm050397. [PMID: 38770680 PMCID: PMC11139035 DOI: 10.1242/dmm.050397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 03/28/2024] [Indexed: 05/22/2024] Open
Abstract
Absence of dystrophin results in muscular weakness, chronic inflammation and cardiomyopathy in Duchenne muscular dystrophy (DMD). Pharmacological corticosteroids are the DMD standard of care; however, they have harsh side effects and unclear molecular benefits. It is uncertain whether signaling by physiological corticosteroids and their receptors plays a modifying role in the natural etiology of DMD. Here, we knocked out the glucocorticoid receptor (GR, encoded by Nr3c1) specifically in myofibers and cardiomyocytes within wild-type and mdx52 mice to dissect its role in muscular dystrophy. Double-knockout mice showed significantly worse phenotypes than mdx52 littermate controls in measures of grip strength, hang time, inflammatory pathology and gene expression. In the heart, GR deletion acted additively with dystrophin loss to exacerbate cardiomyopathy, resulting in enlarged hearts, pathological gene expression and systolic dysfunction, consistent with imbalanced mineralocorticoid signaling. The results show that physiological GR functions provide a protective role during muscular dystrophy, directly contrasting its degenerative role in other disease states. These data provide new insights into corticosteroids in disease pathophysiology and establish a new model to investigate cell-autonomous roles of nuclear receptors and mechanisms of pharmacological corticosteroids.
Collapse
MESH Headings
- Animals
- Mice
- Cardiomyopathies/pathology
- Cardiomyopathies/metabolism
- Dystrophin/metabolism
- Dystrophin/genetics
- Dystrophin/deficiency
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mice, Knockout
- Muscle, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/metabolism
- Myocardium/pathology
- Myocardium/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/drug effects
- Phenotype
- Receptors, Glucocorticoid/metabolism
Collapse
Affiliation(s)
- Trinitee Oliver
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Biology, Howard University, Washington, DC 20059, USA
- Graduate School of Biomedical Sciences, Cedars-Sinai Medical Center, West Hollywood, CA 90048, USA
| | - Nhu Y. Nguyen
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
| | - Christopher B. Tully
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
| | - Nikki M. McCormack
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
| | - Christina M. Sun
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
| | - Alyson A. Fiorillo
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC 20037, USA
- Center for Inherited Muscle Research, Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Christopher R. Heier
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC 20037, USA
- Center for Inherited Muscle Research, Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
19
|
Zhang Q, Li Z, Li Q, Trammell SA, Schmidt MS, Pires KM, Cai J, Zhang Y, Kenny H, Boudina S, Brenner C, Abel ED. Control of NAD + homeostasis by autophagic flux modulates mitochondrial and cardiac function. EMBO J 2024; 43:362-390. [PMID: 38212381 PMCID: PMC10897141 DOI: 10.1038/s44318-023-00009-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 10/31/2023] [Accepted: 11/08/2023] [Indexed: 01/13/2024] Open
Abstract
Impaired autophagy is known to cause mitochondrial dysfunction and heart failure, in part due to altered mitophagy and protein quality control. However, whether additional mechanisms are involved in the development of mitochondrial dysfunction and heart failure in the setting of deficient autophagic flux remains poorly explored. Here, we show that impaired autophagic flux reduces nicotinamide adenine dinucleotide (NAD+) availability in cardiomyocytes. NAD+ deficiency upon autophagic impairment is attributable to the induction of nicotinamide N-methyltransferase (NNMT), which methylates the NAD+ precursor nicotinamide (NAM) to generate N-methyl-nicotinamide (MeNAM). The administration of nicotinamide mononucleotide (NMN) or inhibition of NNMT activity in autophagy-deficient hearts and cardiomyocytes restores NAD+ levels and ameliorates cardiac and mitochondrial dysfunction. Mechanistically, autophagic inhibition causes the accumulation of SQSTM1, which activates NF-κB signaling and promotes NNMT transcription. In summary, we describe a novel mechanism illustrating how autophagic flux maintains mitochondrial and cardiac function by mediating SQSTM1-NF-κB-NNMT signaling and controlling the cellular levels of NAD+.
Collapse
Affiliation(s)
- Quanjiang Zhang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Zhonggang Li
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Department of Human Genetics, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Qiuxia Li
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Samuel Aj Trammell
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Mark S Schmidt
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Karla Maria Pires
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Jinjin Cai
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Yuan Zhang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Helena Kenny
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Sihem Boudina
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, 84112, USA
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Department of Diabetes & Cancer Metabolism, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - E Dale Abel
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
20
|
Silva-Velasco DL, Cervantes-Pérez LG, Sánchez-Mendoza A. ACE inhibitors and their interaction with systems and molecules involved in metabolism. Heliyon 2024; 10:e24655. [PMID: 38298628 PMCID: PMC10828069 DOI: 10.1016/j.heliyon.2024.e24655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
The main function of the renin-angiotensin-aldosterone system (RAAS) is the regulation of blood pressure; therefore, researchers have focused on its study to treat cardiovascular and renal diseases. One of the most widely used treatments derived from the study of RAAS, is the use of angiotensin-converting enzyme inhibitors (ACEi). Since it was discovered, the main target of ACEi has been the cardiovascular and renal systems. However, being the RAAS expressed locally in several specialized tissues and cells such as pneumocytes, hepatocytes, spleenocytes, enterocytes, adipocytes, and neurons the effect of inhibitors has expanded, because it is expected that RAAS has a role in the specific function of those cells. Many chronic degenerative diseases compromise the correct function of those organs, and in most of them, the RAAS is overactivated. Therefore, the use of ACEi must exert a benefit on an impaired system. Accordingly, the objective of this review is to present a brief overview of the cardiovascular and renal actions of ACEi and its effects in organs that are not the classic targets of ACEi that carry on glucose and lipid metabolism.
Collapse
Affiliation(s)
| | - Luz G. Cervantes-Pérez
- Departamento de Farmacología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Alicia Sánchez-Mendoza
- Departamento de Farmacología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
21
|
Wei J, Duan X, Chen J, Zhang D, Xu J, Zhuang J, Wang S. Metabolic adaptations in pressure overload hypertrophic heart. Heart Fail Rev 2024; 29:95-111. [PMID: 37768435 DOI: 10.1007/s10741-023-10353-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
This review article offers a detailed examination of metabolic adaptations in pressure overload hypertrophic hearts, a condition that plays a pivotal role in the progression of heart failure with preserved ejection fraction (HFpEF) to heart failure with reduced ejection fraction (HFrEF). The paper delves into the complex interplay between various metabolic pathways, including glucose metabolism, fatty acid metabolism, branched-chain amino acid metabolism, and ketone body metabolism. In-depth insights into the shifts in substrate utilization, the role of different transporter proteins, and the potential impact of hypoxia-induced injuries are discussed. Furthermore, potential therapeutic targets and strategies that could minimize myocardial injury and promote cardiac recovery in the context of pressure overload hypertrophy (POH) are examined. This work aims to contribute to a better understanding of metabolic adaptations in POH, highlighting the need for further research on potential therapeutic applications.
Collapse
Affiliation(s)
- Jinfeng Wei
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xuefei Duan
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jiaying Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Dengwen Zhang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jindong Xu
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jian Zhuang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Sheng Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Linzhi People's Hospital, Linzhi, Tibet, China.
| |
Collapse
|
22
|
Zhang J, Sandroni PB, Huang W, Gao X, Oswalt L, Schroder MA, Lee S, Shih YYI, Huang HYS, Swigart PM, Myagmar BE, Simpson PC, Rossi JS, Schisler JC, Jensen BC. Cardiomyocyte Alpha-1A Adrenergic Receptors Mitigate Postinfarct Remodeling and Mortality by Constraining Necroptosis. JACC Basic Transl Sci 2024; 9:78-96. [PMID: 38362342 PMCID: PMC10864988 DOI: 10.1016/j.jacbts.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 08/26/2023] [Accepted: 08/29/2023] [Indexed: 02/17/2024]
Abstract
Clinical studies have shown that α1-adrenergic receptor antagonists (α-blockers) are associated with increased heart failure risk. The mechanism underlying that hazard and whether it arises from direct inhibition of cardiomyocyte α1-ARs or from systemic effects remain unclear. To address these issues, we created a mouse with cardiomyocyte-specific deletion of the α1A-AR subtype and found that it experienced 70% mortality within 7 days of myocardial infarction driven, in part, by excessive activation of necroptosis. We also found that patients taking α-blockers at our center were at increased risk of death after myocardial infarction, providing clinical correlation for our translational animal models.
Collapse
Affiliation(s)
- Jiandong Zhang
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Peyton B. Sandroni
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Wei Huang
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Xiaohua Gao
- Department of Epidemiology, University of North Carolina Gillings School of Public Health, Chapel Hill, North Carolina, USA
| | - Leah Oswalt
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Melissa A. Schroder
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - SungHo Lee
- Center for Animal MRI, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Yen-Yu I. Shih
- Center for Animal MRI, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Hsiao-Ying S. Huang
- Mechanical and Aerospace Engineering Department, North Carolina State University, Raleigh, North Carolina, USA
| | - Philip M. Swigart
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Bat E. Myagmar
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Paul C. Simpson
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Joseph S. Rossi
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Jonathan C. Schisler
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Brian C. Jensen
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
23
|
Colosimo S, Mitra SK, Chaudhury T, Marchesini G. Insulin resistance and metabolic flexibility as drivers of liver and cardiac disease in T2DM. Diabetes Res Clin Pract 2023; 206:111016. [PMID: 37979728 DOI: 10.1016/j.diabres.2023.111016] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/15/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Abstract
Metabolic flexibility refers to the ability of tissues to adapt their use of energy sources according to substrate availability and energy demands. This review aims to disentangle the emerging mechanisms through which altered metabolic flexibility and insulin resistance promote NAFLD and heart disease progression. Insulin resistance and metabolic inflexibility are central drivers of hepatic and cardiac diseases in individuals with type 2 diabetes. Both play a critical role in the complex interaction between glucose and lipid metabolism. Disruption of metabolic flexibility results in hyperglycemia and abnormal lipid metabolism, leading to increased accumulation of fat in the liver, contributing to the development and progression of NAFLD. Similarly, insulin resistance affects cardiac glucose metabolism, leading to altered utilization of energy substrates and impaired cardiac function, and influence cardiac lipid metabolism, further exacerbating the progression of heart failure. Regular physical activity promotes metabolic flexibility by increasing energy expenditure and enabling efficient switching between different energy substrates. On the contrary, weight loss achieved through calorie restriction ameliorates insulin sensitivity without improving flexibility. Strategies that mimic the effects of physical exercise, such as pharmacological interventions or targeted lifestyle modifications, show promise in effectively treating both diabetes and NAFLD, finally reducing the risk of advanced liver disease.
Collapse
Affiliation(s)
- Santo Colosimo
- School of Nutrition Science, University of Milan, Milan, Italy
| | - Sandip Kumar Mitra
- Diabetes and Endocrinology Unit, Apollo Gleneagles Hospital, Kolkata, West Bengal, India
| | - Tirthankar Chaudhury
- Diabetes and Endocrinology Unit, Apollo Gleneagles Hospital, Kolkata, West Bengal, India
| | - Giulio Marchesini
- IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Policlinico di Sant'Orsola, Bologna, Italy.
| |
Collapse
|
24
|
Da Dalt L, Cabodevilla AG, Goldberg IJ, Norata GD. Cardiac lipid metabolism, mitochondrial function, and heart failure. Cardiovasc Res 2023; 119:1905-1914. [PMID: 37392421 PMCID: PMC10681665 DOI: 10.1093/cvr/cvad100] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/31/2023] [Accepted: 03/01/2023] [Indexed: 07/03/2023] Open
Abstract
A fine balance between uptake, storage, and the use of high energy fuels, like lipids, is crucial in the homeostasis of different metabolic tissues. Nowhere is this balance more important and more precarious than in the heart. This highly energy-demanding muscle normally oxidizes almost all the available substrates to generate energy, with fatty acids being the preferred source under physiological conditions. In patients with cardiomyopathies and heart failure, changes in the main energetic substrate are observed; these hearts often prefer to utilize glucose rather than oxidizing fatty acids. An imbalance between uptake and oxidation of fatty acid can result in cellular lipid accumulation and cytotoxicity. In this review, we will focus on the sources and uptake pathways used to direct fatty acids to cardiomyocytes. We will then discuss the intracellular machinery used to either store or oxidize these lipids and explain how disruptions in homeostasis can lead to mitochondrial dysfunction and heart failure. Moreover, we will also discuss the role of cholesterol accumulation in cardiomyocytes. Our discussion will attempt to weave in vitro experiments and in vivo data from mice and humans and use several human diseases to illustrate metabolism gone haywire as a cause of or accomplice to cardiac dysfunction.
Collapse
Affiliation(s)
- Lorenzo Da Dalt
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, Milan, Italy
| | - Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, 550 1st Ave., New York, NY, USA
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, 550 1st Ave., New York, NY, USA
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, Milan, Italy
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Via Massimo Gorki 50, Cinisello Balsamo, Italy
| |
Collapse
|
25
|
Sohail S, Akkawi G, Rechter T, Fluitt MB, Ecelbarger CM. Sex Modulates Response to Renal-Tubule-Targeted Insulin Receptor Deletion in Mice. Int J Mol Sci 2023; 24:8056. [PMID: 37175762 PMCID: PMC10178497 DOI: 10.3390/ijms24098056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/19/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Insulin facilitates renal sodium reabsorption and attenuates gluconeogenesis. Sex differences in this regulation have not been well characterized. Using tetracycline-inducible Cre-lox recombination, we knocked out (KO) the insulin receptor (InsR) from the renal tubule in adult male (M) and female (F) mice (C57Bl6 background) with a paired box 8 (PAX8) promoter. Body weights were not affected by the KO, but mean kidney weights were reduced in the KO mice (13 and 3%, in M and F, respectively, relative to wild-type (WT) mice). A microscopic analysis revealed 25 and 19% reductions in the proximal tubule (PT) and cortical collecting duct cell heights, respectively, in KOMs relative to WTMs. The reductions were 5 and 11% for KOFs. Western blotting of renal cortex homogenates showed decreased protein levels for the β and γ subunits of the epithelial sodium channel (ENaC) and the sodium-potassium-2-chloride cotransporter type 2 (NKCC2) in both sexes of KO mice; however, α-ENaC was upregulated in KOMs and downregulated in KOFs. Both sexes of KO mice cleared exogenously administered glucose faster than the WT mice and had lower semi-fasted, anesthetized blood glucose levels. However, KOMs (but not KOFs) demonstrated evidence of enhanced renal gluconeogenesis, including higher levels of renal glucose-6-phosphatase, the PT's production of glucose, post-prandial blood glucose, and plasma insulin, whereas KOFs exhibited downregulation of renal high-capacity sodium glucose cotransporter (SGLT2) and upregulation of SGLT1; these changes appeared to be absent in the KOM. Overall, these findings suggest a sex-differential reliance on intact renal tubular InsR signaling which may be translationally important in type 2 diabetes, obesity, or insulin resistance when renal insulin signaling is reduced.
Collapse
Affiliation(s)
- Soha Sohail
- Department of Medicine, Georgetown University, Washington, DC 20057, USA
| | - Gabriella Akkawi
- Department of Medicine, Georgetown University, Washington, DC 20057, USA
| | - Taylor Rechter
- Department of Medicine, Georgetown University, Washington, DC 20057, USA
| | - Maurice B. Fluitt
- Department of Medicine, Georgetown University, Washington, DC 20057, USA
- Department of Medicine, Howard University, Washington, DC 20059, USA
| | | |
Collapse
|
26
|
Tham YK, Bernardo BC, Claridge B, Yildiz GS, Woon LML, Bond S, Fang H, Ooi JYY, Matsumoto A, Luo J, Tai CMK, Harmawan CA, Kiriazis H, Donner DG, Mellett NA, Abel ED, Khan SA, De Souza DP, Doomun SNE, Liu K, Xiang R, Singh M, Inouye M, Meikle PJ, Weeks KL, Drew BG, Greening DW, McMullen JR. Estrogen receptor alpha deficiency in cardiomyocytes reprograms the heart-derived extracellular vesicle proteome and induces obesity in female mice. NATURE CARDIOVASCULAR RESEARCH 2023; 2:268-289. [PMID: 39196021 DOI: 10.1038/s44161-023-00223-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/30/2023] [Indexed: 08/29/2024]
Abstract
Dysregulation of estrogen receptor alpha (ERα) has been linked with increased metabolic and cardiovascular disease risk. Here, we generate and characterize cardiomyocyte-specific ERα knockout (ERαHKO) mice to assess the role of ERα in the heart. The most striking phenotype was obesity in female ERαHKO but not male ERαHKO mice. Female ERαHKO mice showed cardiac dysfunction, mild glucose and insulin intolerance and reduced ERα gene expression in skeletal muscle and white adipose tissue. Transcriptomic, proteomic, lipidomic and metabolomic analyses revealed evidence of contractile and/or metabolic dysregulation in heart, skeletal muscle and white adipose tissue. We show that heart-derived extracellular vesicles from female ERαHKO mice contain a distinct proteome associated with lipid and metabolic regulation, and have the capacity to metabolically reprogram the target skeletal myocyte proteome with functional impacts on glycolytic capacity and reserve. This multi-omics study uncovers a cardiac-initiated and sex-specific cardiometabolic phenotype regulated by ERα and provides insights into extracellular vesicle-mediated interorgan communication.
Collapse
Affiliation(s)
- Yow Keat Tham
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia
| | - Bianca C Bernardo
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Bethany Claridge
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Gunes S Yildiz
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Simon Bond
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia
| | - Haoyun Fang
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Jenny Y Y Ooi
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia
| | - Aya Matsumoto
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Jieting Luo
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Celeste M K Tai
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Daniel G Donner
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - E Dale Abel
- David Geffen School of Medicine, University of California, Los Angeles, California, CA, USA
| | - Sohaib A Khan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David P De Souza
- Metabolomics Australia, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - Kevin Liu
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Ruidong Xiang
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Manika Singh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Michael Inouye
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - David W Greening
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia.
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, Victoria, Australia.
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia.
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia.
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia.
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia.
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia.
- Central Clinical School, Monash University, Melbourne, Victoria, Australia.
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia.
- Department of Physiology, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
27
|
Gupta A. Cardiac 31P MR spectroscopy: development of the past five decades and future vision-will it be of diagnostic use in clinics? Heart Fail Rev 2023; 28:485-532. [PMID: 36427161 DOI: 10.1007/s10741-022-10287-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
In the past five decades, the use of the magnetic resonance (MR) technique for cardiovascular diseases has engendered much attention and raised the opportunity that the technique could be useful for clinical applications. MR has two arrows in its quiver: One is magnetic resonance imaging (MRI), and the other is magnetic resonance spectroscopy (MRS). Non-invasively, highly advanced MRI provides unique and profound information about the anatomical changes of the heart. Excellently developed MRS provides irreplaceable and insightful evidence of the real-time biochemistry of cardiac metabolism of underpinning diseases. Compared to MRI, which has already been successfully applied in routine clinical practice, MRS still has a long way to travel to be incorporated into routine diagnostics. Considering the exceptional potential of 31P MRS to measure the real-time metabolic changes of energetic molecules qualitatively and quantitatively, how far its powerful technique should be waited before a successful transition from "bench-to-bedside" is enticing. The present review highlights the seminal studies on the chronological development of cardiac 31P MRS in the past five decades and the future vision and challenges to incorporating it for routine diagnostics of cardiovascular disease.
Collapse
Affiliation(s)
- Ashish Gupta
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow, 226014, India.
| |
Collapse
|
28
|
Sheldon C, Kessinger CW, Sun Y, Kontaridis MI, Ma Q, Hammoud SS, Gao Z, Zhang H, Lin Z. Myh6 promoter-driven Cre recombinase excises floxed DNA fragments in a subset of male germline cells. J Mol Cell Cardiol 2023; 175:62-66. [PMID: 36584478 PMCID: PMC9974737 DOI: 10.1016/j.yjmcc.2022.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022]
Abstract
Myh6-Cre transgenic mouse line was known to express Cre recombinase only in the heart. Nevertheless, during breeding Myh6-Cre to Rosa26fstdTom reporter (tdTom) mouse line, we observed that a significant part of their F2 tdTom/+ offspring had tdTom reporter gene universally activated. Our results show that Myh6-Cre transgenic mice have Cre recombinase activity in a subpopulation of the male germline cells, and that Myh6 gene transcripts are enriched in the interstitial Leydig cells and the undifferentiated spermatogonia stem cells. In summary, the current study confirms that the previously known "heart-specific" Myh6 promoter drives Cre expression in the testis.
Collapse
Affiliation(s)
- Caroline Sheldon
- Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - Chase W Kessinger
- Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - Yan Sun
- Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - Maria I Kontaridis
- Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America; Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Qianyi Ma
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Saher Sue Hammoud
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Zibei Gao
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Hui Zhang
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Zhiqiang Lin
- Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America.
| |
Collapse
|
29
|
Binsch C, Barbosa DM, Hansen-Dille G, Hubert M, Hodge SM, Kolasa M, Jeruschke K, Weiß J, Springer C, Gorressen S, Fischer JW, Lienhard M, Herwig R, Börno S, Timmermann B, Cremer AL, Backes H, Chadt A, Al-Hasani H. Deletion of Tbc1d4/As160 abrogates cardiac glucose uptake and increases myocardial damage after ischemia/reperfusion. Cardiovasc Diabetol 2023; 22:17. [PMID: 36707786 PMCID: PMC9881301 DOI: 10.1186/s12933-023-01746-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Type 2 Diabetes mellitus (T2DM) is a major risk factor for cardiovascular disease and associated with poor outcome after myocardial infarction (MI). In T2DM, cardiac metabolic flexibility, i.e. the switch between carbohydrates and lipids as energy source, is disturbed. The RabGTPase-activating protein TBC1D4 represents a crucial regulator of insulin-stimulated glucose uptake in skeletal muscle by controlling glucose transporter GLUT4 translocation. A human loss-of-function mutation in TBC1D4 is associated with impaired glycemic control and elevated T2DM risk. The study's aim was to investigate TBC1D4 function in cardiac substrate metabolism and adaptation to MI. METHODS Cardiac glucose metabolism of male Tbc1d4-deficient (D4KO) and wild type (WT) mice was characterized using in vivo [18F]-FDG PET imaging after glucose injection and ex vivo basal/insulin-stimulated [3H]-2-deoxyglucose uptake in left ventricular (LV) papillary muscle. Mice were subjected to cardiac ischemia/reperfusion (I/R). Heart structure and function were analyzed until 3 weeks post-MI using echocardiography, morphometric and ultrastructural analysis of heart sections, complemented by whole heart transcriptome and protein measurements. RESULTS Tbc1d4-knockout abolished insulin-stimulated glucose uptake in ex vivo LV papillary muscle and in vivo cardiac glucose uptake after glucose injection, accompanied by a marked reduction of GLUT4. Basal cardiac glucose uptake and GLUT1 abundance were not changed compared to WT controls. D4KO mice showed mild impairments in glycemia but normal cardiac function. However, after I/R D4KO mice showed progressively increased LV endsystolic volume and substantially increased infarction area compared to WT controls. Cardiac transcriptome analysis revealed upregulation of the unfolded protein response via ATF4/eIF2α in D4KO mice at baseline. Transmission electron microscopy revealed largely increased extracellular matrix (ECM) area, in line with decreased cardiac expression of matrix metalloproteinases of D4KO mice. CONCLUSIONS TBC1D4 is essential for insulin-stimulated cardiac glucose uptake and metabolic flexibility. Tbc1d4-deficiency results in elevated cardiac endoplasmic reticulum (ER)-stress response, increased deposition of ECM and aggravated cardiac damage following MI. Hence, impaired TBC1D4 signaling contributes to poor outcome after MI.
Collapse
Affiliation(s)
- C. Binsch
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - D. M. Barbosa
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - G. Hansen-Dille
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - M. Hubert
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - S. M. Hodge
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - M. Kolasa
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - K. Jeruschke
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - J. Weiß
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - C. Springer
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - S. Gorressen
- grid.411327.20000 0001 2176 9917Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| | - J. W. Fischer
- grid.411327.20000 0001 2176 9917Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| | - M. Lienhard
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - R. Herwig
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - S. Börno
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - B. Timmermann
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - A. L. Cremer
- grid.418034.a0000 0004 4911 0702Max Planck Institute for Metabolism Research, Cologne, Germany
| | - H. Backes
- grid.418034.a0000 0004 4911 0702Max Planck Institute for Metabolism Research, Cologne, Germany
| | - A. Chadt
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany ,grid.452622.5German Center for Diabetes Research, Partner Düsseldorf, Munich-Neuherberg, Germany
| | - H. Al-Hasani
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany ,grid.452622.5German Center for Diabetes Research, Partner Düsseldorf, Munich-Neuherberg, Germany
| |
Collapse
|
30
|
Tatekoshi Y, Shapiro JS, Liu M, Bianco GM, Tatekoshi A, De Jesus A, Koleini N, Wasserstrom JA, Dillmann WH, Weinberg SE, Ardehali H. [WITHDRAWN] Hexokinase-1 mitochondrial dissociation and protein O-GlcNAcylation drive heart failure with preserved ejection fraction. RESEARCH SQUARE 2023:rs.3.rs-2448086. [PMID: 36747777 PMCID: PMC9901020 DOI: 10.21203/rs.3.rs-2448086/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The authors have requested that this preprint be removed from Research Square.
Collapse
Affiliation(s)
- Yuki Tatekoshi
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL, USA
| | - Jason S Shapiro
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL, USA
| | - Mingyang Liu
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL, USA
| | - George M Bianco
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL, USA
| | - Ayumi Tatekoshi
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL, USA
| | - Adam De Jesus
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL, USA
| | - Navid Koleini
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL, USA
| | - J Andrew Wasserstrom
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL, USA
| | - Wolfgang H Dillmann
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Samuel E Weinberg
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hossein Ardehali
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University, Chicago, IL, USA
| |
Collapse
|
31
|
Luo P, Wang Z, Su C, Li H, Zhang H, Huang Y, Chen W. Chicken GLUT4 undergoes complex alternative splicing events and its expression in striated muscle changes dramatically during development. Poult Sci 2022; 102:102403. [PMID: 36584419 PMCID: PMC9827075 DOI: 10.1016/j.psj.2022.102403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Glucose transporter protein 4 (GLUT4) plays an important role in regulating insulin-mediated glucose homeostasis in mammals. Until now, studies on GLUT4 have focused on mammals mostly, while chicken GLUT4 has been rarely investigated. In this study, chicken GLUT4 mRNA sequences were obtained by combining conventional amplification, 5'- and 3'- rapid amplification of cDNA ends technique (RACE), then bioinformatics analysis on its genomic structure, splicing pattern, subcellular localization prediction and homologous comparisons were carried out. In addition, the distribution of GLUT4 was detected by RT-qPCR in bird's liver and striated muscles (cardiac muscle, pectoralis and leg muscle) at different ages, including embryonic day 14 (E14), E19, 7-day-old (D7), D21 and D49 (n = 3-4). Results showed that chicken GLUT4 gene produced at least 14 transcripts (GenBank accession No: OP491293-OP491306) through alternative splicing and polyadenylation, which predicted encoding 12 types of amino acid (AA) sequences (with length ranged from 65 AA to 519 AA). These proteins contain typical major facilitator superfamily domain of glucose transporters with length variations, sharing a common sequence of 59 AA, and were predicted to have distinct subcellular localization. The dominant transcript (named as T1) consists of 11 exons with an open reading frame being predicted encoding 519 AA. In addition, analyzing on the spatio-temporal expression of chicken GLUT4 showed it dominantly expressed in pectoralis, leg muscles and cardiac muscle, and the mRNA level of chicken GLUT4 dramatically fluctuated with birds' development in cardiac muscle, pectoralis and leg muscles, with the level at D21 significantly higher than that at E14, E19, and D49 (P < 0.05). These data indicated that chicken GLUT4 undergoes complex alternative splicing events, and GLUT4 expression level in striated muscle was subjected to dynamic regulation with birds' development. Results indicate these isoforms may play overlapping and distinct roles in chicken.
Collapse
Affiliation(s)
| | | | | | | | | | - Yanqun Huang
- College of Animal Science, Henan Agricultural University, Zhengzhou, 450000, China.
| | | |
Collapse
|
32
|
Kuhn AR, van Bilsen M. Oncometabolism: A Paradigm for the Metabolic Remodeling of the Failing Heart. Int J Mol Sci 2022; 23:ijms232213902. [PMID: 36430377 PMCID: PMC9699042 DOI: 10.3390/ijms232213902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Heart failure is associated with profound alterations in cardiac intermediary metabolism. One of the prevailing hypotheses is that metabolic remodeling leads to a mismatch between cardiac energy (ATP) production and demand, thereby impairing cardiac function. However, even after decades of research, the relevance of metabolic remodeling in the pathogenesis of heart failure has remained elusive. Here we propose that cardiac metabolic remodeling should be looked upon from more perspectives than the mere production of ATP needed for cardiac contraction and relaxation. Recently, advances in cancer research have revealed that the metabolic rewiring of cancer cells, often coined as oncometabolism, directly impacts cellular phenotype and function. Accordingly, it is well feasible that the rewiring of cardiac cellular metabolism during the development of heart failure serves similar functions. In this review, we reflect on the influence of principal metabolic pathways on cellular phenotype as originally described in cancer cells and discuss their potential relevance for cardiac pathogenesis. We discuss current knowledge of metabolism-driven phenotypical alterations in the different cell types of the heart and evaluate their impact on cardiac pathogenesis and therapy.
Collapse
|
33
|
Murashige D, Jung JW, Neinast MD, Levin MG, Chu Q, Lambert JP, Garbincius JF, Kim B, Hoshino A, Marti-Pamies I, McDaid KS, Shewale SV, Flam E, Yang S, Roberts E, Li L, Morley MP, Bedi KC, Hyman MC, Frankel DS, Margulies KB, Assoian RK, Elrod JW, Jang C, Rabinowitz JD, Arany Z. Extra-cardiac BCAA catabolism lowers blood pressure and protects from heart failure. Cell Metab 2022; 34:1749-1764.e7. [PMID: 36223763 PMCID: PMC9633425 DOI: 10.1016/j.cmet.2022.09.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 06/09/2022] [Accepted: 09/12/2022] [Indexed: 01/24/2023]
Abstract
Pharmacologic activation of branched-chain amino acid (BCAA) catabolism is protective in models of heart failure (HF). How protection occurs remains unclear, although a causative block in cardiac BCAA oxidation is widely assumed. Here, we use in vivo isotope infusions to show that cardiac BCAA oxidation in fact increases, rather than decreases, in HF. Moreover, cardiac-specific activation of BCAA oxidation does not protect from HF even though systemic activation does. Lowering plasma and cardiac BCAAs also fails to confer significant protection, suggesting alternative mechanisms of protection. Surprisingly, activation of BCAA catabolism lowers blood pressure (BP), a known cardioprotective mechanism. BP lowering occurred independently of nitric oxide and reflected vascular resistance to adrenergic constriction. Mendelian randomization studies revealed that elevated plasma BCAAs portend higher BP in humans. Together, these data indicate that BCAA oxidation lowers vascular resistance, perhaps in part explaining cardioprotection in HF that is not mediated directly in cardiomyocytes.
Collapse
Affiliation(s)
- Danielle Murashige
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jae Woo Jung
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael D Neinast
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ 08544, USA
| | - Michael G Levin
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Qingwei Chu
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan P Lambert
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Joanne F Garbincius
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Boa Kim
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Atsushi Hoshino
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ingrid Marti-Pamies
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kendra S McDaid
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Swapnil V Shewale
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily Flam
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven Yang
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Emilia Roberts
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Li Li
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael P Morley
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kenneth C Bedi
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew C Hyman
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David S Frankel
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kenneth B Margulies
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Richard K Assoian
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John W Elrod
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Cholsoon Jang
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ 08544, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Joshua D Rabinowitz
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ 08544, USA
| | - Zoltan Arany
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
34
|
Li X, Wu J, Xu F, Chu C, Li X, Shi X, Zheng W, Wang Z, Jia Y, Xiao W. Use of Ferulic Acid in the Management of Diabetes Mellitus and Its Complications. Molecules 2022; 27:molecules27186010. [PMID: 36144745 PMCID: PMC9503003 DOI: 10.3390/molecules27186010] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 12/06/2022] Open
Abstract
Diabetes mellitus, a metabolic disease mainly characterized by hyperglycemia, is becoming a serious social health problem worldwide with growing prevalence. Many natural compounds have been found to be effective in the prevention and treatment of diabetes, with negligible toxic effects. Ferulic acid (FA), a phenolic compound commonly found in medicinal herbs and the daily diet, was proved to have several pharmacological effects such as antihyperglycemic, antihyperlipidemic and antioxidant actions, which are beneficial to the management of diabetes and its complications. Data from PubMed, EM-BASE, Web of Science and CNKI were searched with the keywords ferulic acid and diabetes mellitus. Finally, 28 articles were identified after literature screening, and the research progress of FA for the management of DM and its complications was summarized in the review, in order to provide references for further research and medical applications of FA.
Collapse
Affiliation(s)
- Xu Li
- Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang 222001, China
| | - Jingxian Wu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fanxing Xu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang 222001, China
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chun Chu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiang Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinyi Shi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wen Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang 222001, China
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
- Correspondence: (Y.J.); (W.X.)
| | - Wei Xiao
- Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang 222001, China
- Correspondence: (Y.J.); (W.X.)
| |
Collapse
|
35
|
Støle TP, Lunde M, Shen X, Martinsen M, Lunde PK, Li J, Lockwood F, Sjaastad I, Louch WE, Aronsen JM, Christensen G, Carlson CR. The female syndecan-4−/− heart has smaller cardiomyocytes, augmented insulin/pSer473-Akt/pSer9-GSK-3β signaling, and lowered SCOP, pThr308-Akt/Akt and GLUT4 levels. Front Cell Dev Biol 2022; 10:908126. [PMID: 36092718 PMCID: PMC9452846 DOI: 10.3389/fcell.2022.908126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: In cardiac muscle, the ubiquitously expressed proteoglycan syndecan-4 is involved in the hypertrophic response to pressure overload. Protein kinase Akt signaling, which is known to regulate hypertrophy, has been found to be reduced in the cardiac muscle of exercised male syndecan-4−/− mice. In contrast, we have recently found that pSer473-Akt signaling is elevated in the skeletal muscle (tibialis anterior, TA) of female syndecan-4−/− mice. To determine if the differences seen in Akt signaling are sex specific, we have presently investigated Akt signaling in the cardiac muscle of sedentary and exercised female syndecan-4−/− mice. To get deeper insight into the female syndecan-4−/− heart, alterations in cardiomyocyte size, a wide variety of different extracellular matrix components, well-known syndecan-4 binding partners and associated signaling pathways have also been investigated.Methods: Left ventricles (LVs) from sedentary and exercise trained female syndecan-4−/− and WT mice were analyzed by immunoblotting and real-time PCR. Cardiomyocyte size and phosphorylated Ser473-Akt were analyzed in isolated adult cardiomyocytes from female syndecan-4−/− and WT mice by confocal imaging. LV and skeletal muscle (TA) from sedentary male syndecan-4−/− and WT mice were immunoblotted with Akt antibodies for comparison. Glucose levels were measured by a glucometer, and fasting blood serum insulin and C-peptide levels were measured by ELISA.Results: Compared to female WT hearts, sedentary female syndecan-4−/− LV cardiomyocytes were smaller and hearts had higher levels of pSer473-Akt and its downstream target pSer9-GSK-3β. The pSer473-Akt inhibitory phosphatase PHLPP1/SCOP was lowered, which may be in response to the elevated serum insulin levels found in the female syndecan-4−/− mice. We also observed lowered levels of pThr308-Akt/Akt and GLUT4 in the female syndecan-4−/− heart and an increased LRP6 level after exercise. Otherwise, few alterations were found. The pThr308-Akt and pSer473-Akt levels were unaltered in the cardiac and skeletal muscles of sedentary male syndecan-4−/− mice.Conclusion: Our data indicate smaller cardiomyocytes, an elevated insulin/pSer473-Akt/pSer9-GSK-3β signaling pathway, and lowered SCOP, pThr308-Akt/Akt and GLUT4 levels in the female syndecan-4−/− heart. In contrast, cardiomyocyte size, and Akt signaling were unaltered in both cardiac and skeletal muscles from male syndecan-4−/− mice, suggesting important sex differences.
Collapse
Affiliation(s)
- Thea Parsberg Støle
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- *Correspondence: Thea Parsberg Støle,
| | - Marianne Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Xin Shen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Marita Martinsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Per Kristian Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Francesca Lockwood
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - William Edward Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Cathrine Rein Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| |
Collapse
|
36
|
Myeloid cell-specific deletion of Capns1 prevents macrophage polarization toward the M1 phenotype and reduces interstitial lung disease in the bleomycin model of systemic sclerosis. Arthritis Res Ther 2022; 24:148. [PMID: 35729674 PMCID: PMC9210712 DOI: 10.1186/s13075-022-02833-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/03/2022] [Indexed: 11/25/2022] Open
Abstract
Background Calpains are a family of calcium-dependent thiol proteases that participate in a wide variety of biological activities. In our recent study, calpain is increased in the sera of scleroderma or systemic sclerosis (SSc). However, the role of calpain in interstitial lung disease (ILD) has not been reported. ILD is a severe complication of SSc, which is the leading cause of death in SSc. The pathogenesis of SSc-related ILD remains incompletely understood. This study investigated the role of myeloid cell calpain in SSc-related ILD. Methods A novel line of mice with myeloid cell-specific deletion of Capns1 (Capns1-ko) was created. SSc-related ILD was induced in Capns1-ko mice and their wild-type littermates by injection 0.l mL of bleomycin (0.4 mg/mL) for 4 weeks. In a separate experiment, a pharmacological inhibitor of calpain PD150606 (Biomol, USA, 3 mg/kg/day, i.p.) daily for 30 days was given to mice after bleomycin injection on daily basis. At the end of the experiment, the animals were killed, skin and lung tissues were collected for the following analysis. Inflammation, fibrosis and calpain activity and cytokines were assessed by histological examinations and ELISA, and immunohistochemical analyses, western blot analysis and Flow cytometry analysis. Results Calpain activities increased in SSc-mouse lungs. Both deletion of Capns1 and administration of PD150606 attenuated dermal sclerosis as evidenced by a reduction of skin thickness and reduced interstitial fibrosis and inflammation in bleomycin model of SSc mice. These effects of reduced calpain expression or activity were associated with prevention of macrophage polarization toward M1 phenotype and consequent reduced production of pro-inflammatory cytokines including TNF-α, IL-12 and IL-23 in lung tissues of Capns1-ko mice with bleomycin model of SSc. Furthermore, inhibition of calpain correlated with an increase in the protein levels of PI3K and phosphorylated AKT1 in lung tissues of the bleomycin model of SSc mice. Conclusions This study for the first time demonstrates that the role of myeloid cell calpain may be promotion of macrophage M1 polarization and pro-inflammatory responses related PI3K/AKT1 signaling. Thus, myeloid cell calpain may be a potential therapeutic target for bleomycin model of SSc-related ILD.
Collapse
|
37
|
Derangements and Reversibility of Energy Metabolism in Failing Hearts Resulting from Volume Overload: Transcriptomics and Metabolomics Analyses. Int J Mol Sci 2022; 23:ijms23126809. [PMID: 35743252 PMCID: PMC9224487 DOI: 10.3390/ijms23126809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 11/29/2022] Open
Abstract
Derangements in cardiac energy metabolism have been shown to contribute to the development of heart failure (HF). This study combined transcriptomics and metabolomics analyses to characterize the changes and reversibility of cardiac energetics in a rat model of cardiac volume overload (VO) with the creation and subsequent closure of aortocaval fistula. Male Sprague–Dawley rats subjected to an aortocaval fistula surgery for 8 and 16 weeks exhibited characteristics of compensated hypertrophy (CH) and HF, respectively, in echocardiographic and hemodynamic studies. Glycolysis was downregulated and directed to the hexosamine biosynthetic pathway (HBP) and O-linked-N-acetylglucosaminylation in the CH phase and was further suppressed during progression to HF. Derangements in fatty acid oxidation were not prominent until the development of HF, as indicated by the accumulation of acylcarnitines. The gene expression and intermediates of the tricarboxylic acid cycle were not significantly altered in this model. Correction of VO largely reversed the differential expression of genes involved in glycolysis, HBP, and fatty acid oxidation in CH but not in HF. Delayed correction of VO in HF resulted in incomplete recovery of defective glycolysis and fatty acid oxidation. These findings may provide insight into the development of innovative strategies to prevent or reverse metabolic derangements in VO-induced HF.
Collapse
|
38
|
Abstract
As a muscular pump that contracts incessantly throughout life, the heart must constantly generate cellular energy to support contractile function and fuel ionic pumps to maintain electrical homeostasis. Thus, mitochondrial metabolism of multiple metabolic substrates such as fatty acids, glucose, ketones, and lactate is essential to ensuring an uninterrupted supply of ATP. Multiple metabolic pathways converge to maintain myocardial energy homeostasis. The regulation of these cardiac metabolic pathways has been intensely studied for many decades. Rapid adaptation of these pathways is essential for mediating the myocardial adaptation to stress, and dysregulation of these pathways contributes to myocardial pathophysiology as occurs in heart failure and in metabolic disorders such as diabetes. The regulation of these pathways reflects the complex interactions of cell-specific regulatory pathways, neurohumoral signals, and changes in substrate availability in the circulation. Significant advances have been made in the ability to study metabolic regulation in the heart, and animal models have played a central role in contributing to this knowledge. This review will summarize metabolic pathways in the heart and describe their contribution to maintaining myocardial contractile function in health and disease. The review will summarize lessons learned from animal models with altered systemic metabolism and those in which specific metabolic regulatory pathways have been genetically altered within the heart. The relationship between intrinsic and extrinsic regulators of cardiac metabolism and the pathophysiology of heart failure and how these have been informed by animal models will be discussed.
Collapse
Affiliation(s)
- Heiko Bugger
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - Nikole J Byrne
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (E.D.A.)
| |
Collapse
|
39
|
Kawakami R, Sunaga H, Iso T, Kaneko R, Koitabashi N, Obokata M, Harada T, Matsui H, Yokoyama T, Kurabayashi M. Ketone body and FGF21 coordinately regulate fasting-induced oxidative stress response in the heart. Sci Rep 2022; 12:7338. [PMID: 35513524 PMCID: PMC9072431 DOI: 10.1038/s41598-022-10993-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 04/12/2022] [Indexed: 11/09/2022] Open
Abstract
Ketone body β-hydroxybutyrate (βOHB) and fibroblast growth factor-21 (FGF21) have been proposed to mediate systemic metabolic response to fasting. However, it remains elusive about the signaling elicited by ketone and FGF21 in the heart. Stimulation of neonatal rat cardiomyocytes with βOHB and FGF21 induced peroxisome proliferator-activated receptor α (PPARα) and PGC1α expression along with the phosphorylation of LKB1 and AMPK. βOHB and FGF21 induced transcription of peroxisome proliferator-activated receptor response element (PPRE)-containing genes through an activation of PPARα. Additionally, βOHB and FGF21 induced the expression of Nrf2, a master regulator for oxidative stress response, and catalase and Ucp2 genes. We evaluated the oxidative stress response gene expression after 24 h fast in global Fgf21-null (Fgf21-/-) mice, cardiomyocyte-specific FGF21-null (cmFgf21-/-) mice, wild-type (WT), and Fgf21fl/fl littermates. Fgf21-/- mice but not cmFgf21-/- mice had unexpectedly higher serum βOHB levels, and higher expression levels of PPARα and oxidative stress response genes than WT mice or Fgf21fl/fl littermates. Notably, expression levels of oxidative stress response genes were significantly correlated with serum βOHB and PGC1α levels in both WT and Fgf21-/- mice. These findings suggest that fasting-induced βOHB and circulating FGF21 coordinately regulate oxidative stress response gene expression in the heart.
Collapse
Affiliation(s)
- Ryo Kawakami
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroaki Sunaga
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan.,Center for Liberal Arts and Sciences, Ashikaga University, 268-1 Omae-machi, Ashikaga, Tochigi, 326-8558, Japan
| | - Tatsuya Iso
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Ryosuke Kaneko
- Bioresource Center, Gunma University, Graduate School of Medicine, Maebashi, Gunma, Japan.,Osaka University, Graduate School of Frontier Biosciences, 1-3 Yamadaoka, Suita, Osaka, Japan
| | - Norimichi Koitabashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Masaru Obokata
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Tomonari Harada
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroki Matsui
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Gunma, Japan
| | - Tomoyuki Yokoyama
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Gunma, Japan
| | - Masahiko Kurabayashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| |
Collapse
|
40
|
Murugasamy K, Munjal A, Sundaresan NR. Emerging Roles of SIRT3 in Cardiac Metabolism. Front Cardiovasc Med 2022; 9:850340. [PMID: 35369299 PMCID: PMC8971545 DOI: 10.3389/fcvm.2022.850340] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/31/2022] [Indexed: 12/17/2022] Open
Abstract
The heart is a highly metabolically active organ that predominantly utilizes fatty acids as an energy substrate. The heart also derives some part of its energy by oxidation of other substrates, including glucose, lactose, amino acids and ketones. The critical feature of cardiac pathology is metabolic remodeling and loss of metabolic flexibility. Sirtuin 3 (SIRT3) is one of the seven mammalian sirtuins (SIRT1 to SIRT7), with NAD+ dependent deacetylase activity. SIRT3 is expressed in high levels in healthy hearts but downregulated in the aged or diseased hearts. Experimental evidence shows that increasing SIRT3 levels or activity can ameliorate several cardiac pathologies. The primary deacetylation targets of SIRT3 are mitochondrial proteins, most of which are involved in energy metabolism. Thus, SIRT3 improves cardiac health by modulating cardiac energetics. In this review, we discuss the essential role of SIRT3 in regulating cardiac metabolism in the context of physiology and pathology. Specifically, we summarize the recent advancements that emphasize the critical role of SIRT3 as a master regulator of cardiac metabolism. We also present a comprehensive view of all known activators of SIRT3, and elaborate on their therapeutic potential to ameliorate energetic abnormalities in various cardiac pathologies.
Collapse
|
41
|
Wang TN, Hu XG, Chen GX. Uses of knockout, knockdown, and transgenic models in the studies of glucose transporter 4. World J Meta-Anal 2022; 10:1-11. [DOI: 10.13105/wjma.v10.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/10/2021] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
|
42
|
Harris MP, Zhang QJ, Cochran CT, Ponce J, Alexander S, Kronemberger A, Fuqua JD, Zhang Y, Fattal R, Harper T, Murry ML, Grueter CE, Abel ED, Lira VA. Perinatal versus adult loss of ULK1 and ULK2 distinctly influences cardiac autophagy and function. Autophagy 2022; 18:2161-2177. [PMID: 35104184 PMCID: PMC9466614 DOI: 10.1080/15548627.2021.2022289] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Impairments in macroautophagy/autophagy, which degrades dysfunctional organelles as well as long-lived and aggregate proteins, are associated with several cardiomyopathies; however, the regulation of cardiac autophagy remains insufficiently understood. In this regard, ULK1 and ULK2 are thought to play primarily redundant roles in autophagy initiation, but whether their function is developmentally determined, potentially having an impact on cardiac integrity and function remains unknown. Here, we demonstrate that perinatal loss of ULK1 or ULK2 in cardiomyocytes (cU1-KO and cU2-KO mice, respectively) enhances basal autophagy without altering autophagy machinery content while preserving cardiac function. This increased basal autophagy is dependent on the remaining ULK protein given that perinatal loss of both ULK1 and ULK2 in cU1/2-DKO mice impaired autophagy causing age-related cardiomyopathy and reduced survival. Conversely, adult loss of cardiac ULK1, but not of ULK2 (i.e., icU1-KO and icU2-KO mice, respectively), led to a rapidly developing cardiomyopathy, heart failure and early death. icU1-KO mice had impaired autophagy with robust deficits in mitochondrial respiration and ATP synthesis. Trehalose ameliorated autophagy impairments in icU1-KO hearts but did not delay cardiac dysfunction suggesting that ULK1 plays other critical, autophagy-independent, functions in the adult heart. Collectively, these results indicate that cardiac ULK1 and ULK2 are functionally redundant in the developing heart, while ULK1 assumes a more unique, prominent role in the adult heart. Abbreviations: ATG4: autophagy related 4, cysteine peptidase; ATG5: autophagy related 5; ATG7: autophagy related 7; ATG9: autophagy related 9; ATG13: autophagy related 13; CYCS: Cytochrome C; DNM1L, dynamin 1-like; MAP1LC3A: microtubule-associated protein 1 light chain 3 alpha; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; MFN1: mitofusin 1; MFN2: mitofusin 2; MT-CO1: mitochondrially encoded cytochrome c oxidase I; MYH: myosin, heavy polypeptide; NBR1: NBR1 autophagy cargo receptor; NDUFA9: NADH:ubiquinone oxidoreductase subunit A9; OPA1: OPA1, mitochondrial dynamin like GTPase; PPARGC1A, peroxisome proliferator activated receptor, gamma, coactivator 1 alpha; SDHA: succinate dehydrogenase complex, subunit A, flavoprotein (Fp); SQSTM1: sequestosome 1; ULK1: unc-51 like kinase 1; ULK2: unc-51 like kinase 2; UQCRC1: ubiquinol-cytochrome c reductase core protein 1
Collapse
Affiliation(s)
- Matthew P Harris
- Department of Health & Human Physiology, The University of Iowa, Iowa City, IA, USA
| | - Quan J Zhang
- Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA.,Fraternal Order of Eagles Diabetes Research Center, The University of Iowa, Iowa City, IA, USA.,Abboud Cardiovascular Research Center, The University of Iowa, Iowa City, IA, USA
| | - Cole T Cochran
- Department of Health & Human Physiology, The University of Iowa, Iowa City, IA, USA
| | - Jessica Ponce
- Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA
| | - Sean Alexander
- Department of Health & Human Physiology, The University of Iowa, Iowa City, IA, USA
| | - Ana Kronemberger
- Department of Health & Human Physiology, The University of Iowa, Iowa City, IA, USA
| | - Jordan D Fuqua
- Department of Health & Human Physiology, The University of Iowa, Iowa City, IA, USA
| | - Yuan Zhang
- Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA
| | - Ranan Fattal
- Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA
| | - Tyler Harper
- Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA
| | - Matthew L Murry
- Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA
| | - Chad E Grueter
- Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA.,Fraternal Order of Eagles Diabetes Research Center, The University of Iowa, Iowa City, IA, USA.,Abboud Cardiovascular Research Center, The University of Iowa, Iowa City, IA, USA.,Obesity Research and Education Initiative, The University of Iowa, Iowa City, IA, USA.,Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA
| | - E Dale Abel
- Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA.,Fraternal Order of Eagles Diabetes Research Center, The University of Iowa, Iowa City, IA, USA.,Abboud Cardiovascular Research Center, The University of Iowa, Iowa City, IA, USA.,Obesity Research and Education Initiative, The University of Iowa, Iowa City, IA, USA.,Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA
| | - Vitor A Lira
- Department of Health & Human Physiology, The University of Iowa, Iowa City, IA, USA.,Fraternal Order of Eagles Diabetes Research Center, The University of Iowa, Iowa City, IA, USA.,Abboud Cardiovascular Research Center, The University of Iowa, Iowa City, IA, USA.,Obesity Research and Education Initiative, The University of Iowa, Iowa City, IA, USA.,Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
43
|
Jia D, Zhang J, Liu X, Andersen JP, Tian Z, Nie J, Shi Y. Insulin Resistance in Skeletal Muscle Selectively Protects the Heart in Response to Metabolic Stress. Diabetes 2021; 70:2333-2343. [PMID: 34244238 PMCID: PMC8576508 DOI: 10.2337/db20-1212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 07/02/2021] [Indexed: 11/13/2022]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are the leading causes of cardiovascular morbidity and mortality. Although insulin resistance is believed to underlie these disorders, anecdotal evidence contradicts this common belief. Accordingly, obese patients with cardiovascular disease have better prognoses relative to leaner patients with the same diagnoses, whereas treatment of T2DM patients with thiazolidinedione, one of the popular insulin-sensitizer drugs, significantly increases the risk of heart failure. Using mice with skeletal musclespecific ablation of the insulin receptor gene (MIRKO), we addressed this paradox by demonstrating that insulin signaling in skeletal muscles specifically mediated cross talk with the heart, but not other metabolic tissues, to prevent cardiac dysfunction in response to metabolic stress. Despite severe hyperinsulinemia and aggregating obesity, MIRKO mice were protected from myocardial insulin resistance, mitochondrial dysfunction, and metabolic reprogramming in response to diet-induced obesity. Consequently, the MIRKO mice were also protected from myocardial inflammation, cardiomyopathy, and left ventricle dysfunction. Together, our findings suggest that insulin resistance in skeletal muscle functions as a double-edged sword in metabolic diseases.
Collapse
Affiliation(s)
- Dandan Jia
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, Shaanxi, China
- Department of Pharmacology, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Jun Zhang
- Department of Pharmacology, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Xueling Liu
- Department of Pharmacology, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - John-Paul Andersen
- Department of Pharmacology, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Jia Nie
- Department of Pharmacology, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Yuguang Shi
- Department of Pharmacology, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX
| |
Collapse
|
44
|
Dong S, Qian L, Cheng Z, Chen C, Wang K, Hu S, Zhang X, Wu T. Lactate and Myocardiac Energy Metabolism. Front Physiol 2021; 12:715081. [PMID: 34483967 PMCID: PMC8415870 DOI: 10.3389/fphys.2021.715081] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/29/2021] [Indexed: 12/05/2022] Open
Abstract
The myocardium is capable of utilizing different energy substrates, which is referred to as "metabolic flexibility." This process assures ATP production from fatty acids, glucose, lactate, amino acids, and ketones, in the face of varying metabolic contexts. In the normal physiological state, the oxidation of fatty acids contributes to approximately 60% of energy required, and the oxidation of other substrates provides the rest. The accumulation of lactate in ischemic and hypoxic tissues has traditionally be considered as a by-product, and of little utility. However, recent evidence suggests that lactate may represent an important fuel for the myocardium during exercise or myocadiac stress. This new paradigm drives increasing interest in understanding its role in cardiac metabolism under both physiological and pathological conditions. In recent years, blood lactate has been regarded as a signal of stress in cardiac disease, linking to prognosis in patients with myocardial ischemia or heart failure. In this review, we discuss the importance of lactate as an energy source and its relevance to the progression and management of heart diseases.
Collapse
Affiliation(s)
- Shuohui Dong
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Linhui Qian
- Department of Colorectal and Anal Surgery, Feicheng Hospital Affiliated to Shandong First Medical University, Feicheng, China
| | - Zhiqiang Cheng
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Chang Chen
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Kexin Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Sanyuan Hu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Xiang Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Tongzhi Wu
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
45
|
Abstract
Insulin receptors are highly expressed in the heart and vasculature. Insulin signaling regulates cardiac growth, survival, substrate uptake, utilization, and mitochondrial metabolism. Insulin signaling modulates the cardiac responses to physiological and pathological stressors. Altered insulin signaling in the heart may contribute to the pathophysiology of ventricular remodeling and heart failure progression. Myocardial insulin signaling adapts rapidly to changes in the systemic metabolic milieu. What may initially represent an adaptation to protect the heart from carbotoxicity may contribute to amplifying the risk of heart failure in obesity and diabetes. This review article presents the multiple roles of insulin signaling in cardiac physiology and pathology and discusses the potential therapeutic consequences of modulating myocardial insulin signaling.
Collapse
Affiliation(s)
- E Dale Abel
- Division of Endocrinology, Metabolism and Diabetes and Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
46
|
Liang Y, Lyon RC, Pellman J, Bradford WH, Lange S, Bogomolovas J, Dalton ND, Gu Y, Bobar M, Lee MH, Iwakuma T, Nigam V, Asimaki A, Scheinman M, Peterson KL, Sheikh F. Desmosomal COP9 regulates proteome degradation in arrhythmogenic right ventricular dysplasia/cardiomyopathy. J Clin Invest 2021; 131:137689. [PMID: 33857019 PMCID: PMC8159691 DOI: 10.1172/jci137689] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/14/2021] [Indexed: 12/28/2022] Open
Abstract
Dysregulated protein degradative pathways are increasingly recognized as mediators of human disease. This mechanism may have particular relevance to desmosomal proteins that play critical structural roles in both tissue architecture and cell-cell communication, as destabilization/breakdown of the desmosomal proteome is a hallmark of genetic-based desmosomal-targeted diseases, such as the cardiac disease arrhythmogenic right ventricular dysplasia/cardiomyopathy (ARVD/C). However, no information exists on whether there are resident proteins that regulate desmosomal proteome homeostasis. Here, we uncovered a cardiac constitutive photomorphogenesis 9 (COP9) desmosomal resident protein complex, composed of subunit 6 of the COP9 signalosome (CSN6), that enzymatically restricted neddylation and targeted desmosomal proteome degradation. CSN6 binding, localization, levels, and function were affected in hearts of classic mouse and human models of ARVD/C affected by desmosomal loss and mutations, respectively. Loss of desmosomal proteome degradation control due to junctional reduction/loss of CSN6 and human desmosomal mutations destabilizing junctional CSN6 were also sufficient to trigger ARVD/C in mice. We identified a desmosomal resident regulatory complex that restricted desmosomal proteome degradation and disease.
Collapse
Affiliation(s)
- Yan Liang
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Robert C. Lyon
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Jason Pellman
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - William H. Bradford
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Stephan Lange
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Institute of Medicine, Department of Molecular and Clinical Medicine and Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Julius Bogomolovas
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Nancy D. Dalton
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Yusu Gu
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Marcus Bobar
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Mong-Hong Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Vishal Nigam
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- Department of Pediatrics, Seattle Children’s Research Institute and University of Washington, Seattle, Washington, USA
| | - Angeliki Asimaki
- Cardiology Clinical Academic Group, St. George’s University of London, London, United Kingdom
| | - Melvin Scheinman
- Department of Medicine, Cardiac Electrophysiology Section, University of California San Francisco, San Francisco, California, USA
| | - Kirk L. Peterson
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Farah Sheikh
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
47
|
Perez DM. Targeting Adrenergic Receptors in Metabolic Therapies for Heart Failure. Int J Mol Sci 2021; 22:5783. [PMID: 34071350 PMCID: PMC8198887 DOI: 10.3390/ijms22115783] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
The heart has a reduced capacity to generate sufficient energy when failing, resulting in an energy-starved condition with diminished functions. Studies have identified numerous changes in metabolic pathways in the failing heart that result in reduced oxidation of both glucose and fatty acid substrates, defects in mitochondrial functions and oxidative phosphorylation, and inefficient substrate utilization for the ATP that is produced. Recent early-phase clinical studies indicate that inhibitors of fatty acid oxidation and antioxidants that target the mitochondria may improve heart function during failure by increasing compensatory glucose oxidation. Adrenergic receptors (α1 and β) are a key sympathetic nervous system regulator that controls cardiac function. β-AR blockers are an established treatment for heart failure and α1A-AR agonists have potential therapeutic benefit. Besides regulating inotropy and chronotropy, α1- and β-adrenergic receptors also regulate metabolic functions in the heart that underlie many cardiac benefits. This review will highlight recent studies that describe how adrenergic receptor-mediated metabolic pathways may be able to restore cardiac energetics to non-failing levels that may offer promising therapeutic strategies.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
48
|
Abstract
Alterations in cardiac energy metabolism contribute to the severity of heart failure. However, the energy metabolic changes that occur in heart failure are complex and are dependent not only on the severity and type of heart failure present but also on the co-existence of common comorbidities such as obesity and type 2 diabetes. The failing heart faces an energy deficit, primarily because of a decrease in mitochondrial oxidative capacity. This is partly compensated for by an increase in ATP production from glycolysis. The relative contribution of the different fuels for mitochondrial ATP production also changes, including a decrease in glucose and amino acid oxidation, and an increase in ketone oxidation. The oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in heart failure associated with diabetes and obesity, myocardial fatty acid oxidation increases, while in heart failure associated with hypertension or ischemia, myocardial fatty acid oxidation decreases. Combined, these energy metabolic changes result in the failing heart becoming less efficient (ie, a decrease in cardiac work/O2 consumed). The alterations in both glycolysis and mitochondrial oxidative metabolism in the failing heart are due to both transcriptional changes in key enzymes involved in these metabolic pathways, as well as alterations in NAD redox state (NAD+ and nicotinamide adenine dinucleotide levels) and metabolite signaling that contribute to posttranslational epigenetic changes in the control of expression of genes encoding energy metabolic enzymes. Alterations in the fate of glucose, beyond flux through glycolysis or glucose oxidation, also contribute to the pathology of heart failure. Of importance, pharmacological targeting of the energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac efficiency, decreasing the energy deficit and improving cardiac function in the failing heart.
Collapse
Affiliation(s)
- Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Rong Tian
- Mitochondria and Metabolism Center, University of Washington, Seattle (R.T.)
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.)
| | - E Dale Abel
- Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City (E.D.A.).,Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City (E.D.A.)
| |
Collapse
|
49
|
McMillin SL, Stanley EC, Weyrauch LA, Brault JJ, Kahn BB, Witczak CA. Insulin Resistance Is Not Sustained Following Denervation in Glycolytic Skeletal Muscle. Int J Mol Sci 2021; 22:4913. [PMID: 34066429 PMCID: PMC8125496 DOI: 10.3390/ijms22094913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 02/03/2023] Open
Abstract
Denervation rapidly induces insulin resistance (i.e., impairments in insulin-stimulated glucose uptake and signaling proteins) in skeletal muscle. Surprisingly, whether this metabolic derangement is long-lasting is presently not clear. The main goal of this study was to determine if insulin resistance is sustained in both oxidative soleus and glycolytic extensor digitorum longus (EDL) muscles following long-term (28 days) denervation. Mouse hindlimb muscles were denervated via unilateral sciatic nerve resection. Both soleus and EDL muscles atrophied ~40%. Strikingly, while denervation impaired submaximal insulin-stimulated [3H]-2-deoxyglucose uptake ~30% in the soleus, it enhanced submaximal (~120%) and maximal (~160%) insulin-stimulated glucose uptake in the EDL. To assess possible mechanism(s), immunoblots were performed. Denervation did not consistently alter insulin signaling (e.g., p-Akt (Thr308):Akt; p-TBC1D1 [phospho-Akt substrate (PAS)]:TBC1D1; or p-TBC1D4 (PAS):TBC1D4) in either muscle. However, denervation decreased glucose transporter 4 (GLUT4) levels ~65% in the soleus but increased them ~90% in the EDL. To assess the contribution of GLUT4 to the enhanced EDL muscle glucose uptake, muscle-specific GLUT4 knockout mice were examined. Loss of GLUT4 prevented the denervation-induced increase in insulin-stimulated glucose uptake. In conclusion, the denervation results sustained insulin resistance in the soleus but enhanced insulin sensitivity in the EDL due to increased GLUT4 protein levels.
Collapse
Affiliation(s)
- Shawna L. McMillin
- Department of Kinesiology, East Carolina University, Greenville, NC 27858, USA; (S.L.M.); (E.C.S.); (L.A.W.); (J.J.B.)
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Erin C. Stanley
- Department of Kinesiology, East Carolina University, Greenville, NC 27858, USA; (S.L.M.); (E.C.S.); (L.A.W.); (J.J.B.)
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Luke A. Weyrauch
- Department of Kinesiology, East Carolina University, Greenville, NC 27858, USA; (S.L.M.); (E.C.S.); (L.A.W.); (J.J.B.)
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Jeffrey J. Brault
- Department of Kinesiology, East Carolina University, Greenville, NC 27858, USA; (S.L.M.); (E.C.S.); (L.A.W.); (J.J.B.)
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, NC 27834, USA
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Barbara B. Kahn
- Division of Endocrinology, Diabetes & Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA;
| | - Carol A. Witczak
- Department of Kinesiology, East Carolina University, Greenville, NC 27858, USA; (S.L.M.); (E.C.S.); (L.A.W.); (J.J.B.)
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, NC 27834, USA
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
50
|
Fajardo VM, Feng I, Chen BY, Perez-Ramirez CA, Shi B, Clark P, Tian R, Lien CL, Pellegrini M, Christofk H, Nakano H, Nakano A. GLUT1 overexpression enhances glucose metabolism and promotes neonatal heart regeneration. Sci Rep 2021; 11:8669. [PMID: 33883682 PMCID: PMC8060418 DOI: 10.1038/s41598-021-88159-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/09/2021] [Indexed: 01/10/2023] Open
Abstract
The mammalian heart switches its main metabolic substrate from glucose to fatty acids shortly after birth. This metabolic switch coincides with the loss of regenerative capacity in the heart. However, it is unknown whether glucose metabolism regulates heart regeneration. Here, we report that glucose metabolism is a determinant of regenerative capacity in the neonatal mammalian heart. Cardiac-specific overexpression of Glut1, the embryonic form of constitutively active glucose transporter, resulted in an increase in glucose uptake and concomitant accumulation of glycogen storage in postnatal heart. Upon cryoinjury, Glut1 transgenic hearts showed higher regenerative capacity with less fibrosis than non-transgenic control hearts. Interestingly, flow cytometry analysis revealed two distinct populations of ventricular cardiomyocytes: Tnnt2-high and Tnnt2-low cardiomyocytes, the latter of which showed significantly higher mitotic activity in response to high intracellular glucose in Glut1 transgenic hearts. Metabolic profiling shows that Glut1-transgenic hearts have a significant increase in the glucose metabolites including nucleotides upon injury. Inhibition of the nucleotide biosynthesis abrogated the regenerative advantage of high intra-cardiomyocyte glucose level, suggesting that the glucose enhances the cardiomyocyte regeneration through the supply of nucleotides. Our data suggest that the increase in glucose metabolism promotes cardiac regeneration in neonatal mouse heart.
Collapse
Affiliation(s)
- Viviana M Fajardo
- Division of Neonatology, Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Iris Feng
- Department of Molecular, Cell, Developmental Biology, School of Life Science, University of California Los Angeles, Los Angeles, CA, USA
| | - Bao Ying Chen
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, CA, USA
| | - Cesar A Perez-Ramirez
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Baochen Shi
- Department of Molecular, Cell, Developmental Biology, School of Life Science, University of California Los Angeles, Los Angeles, CA, USA
| | - Peter Clark
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, CA, USA
| | - Rong Tian
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Ching-Ling Lien
- The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Surgery, University of Southern California, Los Angeles, CA, USA
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell, Developmental Biology, School of Life Science, University of California Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
| | - Heather Christofk
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
| | - Haruko Nakano
- Department of Molecular, Cell, Developmental Biology, School of Life Science, University of California Los Angeles, Los Angeles, CA, USA
| | - Atsushi Nakano
- Department of Molecular, Cell, Developmental Biology, School of Life Science, University of California Los Angeles, Los Angeles, CA, USA.
- Division of Cardiology, Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|