1
|
Comella F, Aragón-Herrera A, Pirozzi C, Feijóo-Bandin S, Lama A, Opallo N, Melini S, Del Piano F, Gualillo O, Meli R, Mattace Raso G, Lago F. Oleoylethanolamide mitigates cardiometabolic disruption secondary to obesity induced by high-fat diet in mice. Life Sci 2024; 359:123226. [PMID: 39515418 DOI: 10.1016/j.lfs.2024.123226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/24/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Chronic lipid overnutrition has been demonstrated to promote cardiac dysfunction resulting from metabolic derangement, inflammation, and fibrosis. Oleoylethanolamide (OEA), an endogenous peroxisome proliferator activating receptor (PPAR)-α agonist, has been extensively studied for its metabolic properties. The aim of this study was to determine if OEA has beneficial effects on high-fat diet (HFD)-induced cardiac disruption in obese mice, focusing on the underlying pathological mechanisms. OEA treatment restores the metabolic pattern, improving serum glycaemic and lipid profile. OEA also reduces heart weight and serum creatine kinase-myocardial band (CK-MB), a marker of cardiac damage. Accordingly, OEA modulates cardiac metabolism, increasing insulin signaling and reducing lipid accumulation. OEA increases AMPK and AKT phosphorylation, converging in the rise of AS160 activation and glucose transporter (GLUT)4 protein level. Moreover, OEA reduces the transcription of the cardiac fatty acid transporter CD36 and fatty acid synthase and increases PPAR-α mRNA levels. Adiponectin and meteorite-like protein transcription levels were significantly reduced by OEA in HFD mice, as well as those of inflammatory cytokines and pro-fibrotic markers. An increased autophagic process was also shown, contributing to OEA's cardioprotective effects. Metabolomic analyses of cardiac tissue revealed the modulation of different lipids, including triglycerides, glycerophospholipids and sphingomyelins by OEA treatment. In vitro experiments on HL-1 cardiomyocytes showed OEA's capability in reducing inflammation and fibrosis following palmitate challenge, demonstrating a direct activity of OEA on cardiac cells, mainly mediated by PPAR-α activation. Our results indicate OEA as a potential therapeutic to restrain cardiac damage associated with metabolic disorders.
Collapse
Affiliation(s)
- Federica Comella
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Alana Aragón-Herrera
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Área Sanitaria de Santiago de Compostela y Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Claudio Pirozzi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Sandra Feijóo-Bandin
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Área Sanitaria de Santiago de Compostela y Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Adriano Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Nicola Opallo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Stefania Melini
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Filomena Del Piano
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II, 80100 Naples, Italy
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Group (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Building C, Travesía da Choupana SIN, Santiago de Compostela 15706, Spain; International PhD School, University of Santiago de Compostela (EDIUS), Santiago de Compostela 15706, Spain
| | - Rosaria Meli
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppina Mattace Raso
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy.
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Área Sanitaria de Santiago de Compostela y Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
2
|
Achter JS, Vega ET, Sorrentino A, Kahnert K, Galsgaard KD, Hernandez-Varas P, Wierer M, Holst JJ, Wojtaszewski JFP, Mills RW, Kjøbsted R, Lundby A. In-depth phosphoproteomic profiling of the insulin signaling response in heart tissue and cardiomyocytes unveils canonical and specialized regulation. Cardiovasc Diabetol 2024; 23:258. [PMID: 39026321 PMCID: PMC11264841 DOI: 10.1186/s12933-024-02338-4] [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: 03/12/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Insulin signaling regulates cardiac substrate utilization and is implicated in physiological adaptations of the heart. Alterations in the signaling response within the heart are believed to contribute to pathological conditions such as type-2 diabetes and heart failure. While extensively investigated in several metabolic organs using phosphoproteomic strategies, the signaling response elicited in cardiac tissue in general, and specifically in the specialized cardiomyocytes, has not yet been investigated to the same extent. METHODS Insulin or vehicle was administered to male C57BL6/JRj mice via intravenous injection into the vena cava. Ventricular tissue was extracted and subjected to quantitative phosphoproteomics analysis to evaluate the insulin signaling response. To delineate the cardiomyocyte-specific response and investigate the role of Tbc1d4 in insulin signal transduction, cardiomyocytes from the hearts of cardiac and skeletal muscle-specific Tbc1d4 knockout mice, as well as from wildtype littermates, were studied. The phosphoproteomic studies involved isobaric peptide labeling with Tandem Mass Tags (TMT), enrichment for phosphorylated peptides, fractionation via micro-flow reversed-phase liquid chromatography, and high-resolution mass spectrometry measurements. RESULTS We quantified 10,399 phosphorylated peptides from ventricular tissue and 12,739 from isolated cardiomyocytes, localizing to 3,232 and 3,128 unique proteins, respectively. In cardiac tissue, we identified 84 insulin-regulated phosphorylation events, including sites on the Insulin Receptor (InsrY1351, Y1175, Y1179, Y1180) itself as well as the Insulin receptor substrate protein 1 (Irs1S522, S526). Predicted kinases with increased activity in response to insulin stimulation included Rps6kb1, Akt1 and Mtor. Tbc1d4 emerged as a major phosphorylation target in cardiomyocytes. Despite limited impact on the global phosphorylation landscape, Tbc1d4 deficiency in cardiomyocytes attenuated insulin-induced Glut4 translocation and induced protein remodeling. We observed 15 proteins significantly regulated upon knockout of Tbc1d4. While Glut4 exhibited decreased protein abundance consequent to Tbc1d4-deficiency, Txnip levels were notably increased. Stimulation of wildtype cardiomyocytes with insulin led to the regulation of 262 significant phosphorylation events, predicted to be regulated by kinases such as Akt1, Mtor, Akt2, and Insr. In cardiomyocytes, the canonical insulin signaling response is elicited in addition to regulation on specialized cardiomyocyte proteins, such as Kcnj11Y12 and DspS2597. Details of all phosphorylation sites are provided. CONCLUSION We present a first global outline of the insulin-induced phosphorylation signaling response in heart tissue and in isolated adult cardiomyocytes, detailing the specific residues with changed phosphorylation abundances. Our study marks an important step towards understanding the role of insulin signaling in cardiac diseases linked to insulin resistance.
Collapse
Affiliation(s)
- Jonathan Samuel Achter
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Estefania Torres Vega
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Sorrentino
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Konstantin Kahnert
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Douglas Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pablo Hernandez-Varas
- Core Facility for Integrated Microscopy, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Wierer
- Proteomics Research Infrastructure, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen Frank Pind Wojtaszewski
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Robert William Mills
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Kjøbsted
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
3
|
Xing X, Sun Q, Wang R, Wang Y, Wang R. Impacts of glutamate, an exercise-responsive metabolite on insulin signaling. Life Sci 2024; 341:122471. [PMID: 38301875 DOI: 10.1016/j.lfs.2024.122471] [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: 11/28/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/03/2024]
Abstract
AIMS Disruption of the insulin signaling pathway leads to insulin resistance (IR). IR is characterized by impaired glucose and lipid metabolism. Elevated levels of circulating glutamate are correlated with metabolic indicators and may potentially predict the onset of metabolic diseases. Glutamate receptor antagonists have significantly enhanced insulin sensitivity, and improved glucose and lipid metabolism. Exercise is a well-known strategy to combat IR. The aims of our narrative review are to summarize preclinical and clinical findings to show the correlations between circulating glutamate levels, IR and metabolic diseases, discuss the causal role of excessive glutamate in IR and metabolic disturbance, and present an overview of the exercise-induced alteration in circulating glutamate levels. MATERIALS AND METHODS A literature search was conducted to identify studies on glutamate, insulin signaling, and exercise in the PubMed database. The search covered articles published from December 1955 to January 2024, using the search terms of "glutamate", "glutamic acid", "insulin signaling", "insulin resistance", "insulin sensitivity", "exercise", and "physical activity". KEY FINDINGS Elevated levels of circulating glutamate are correlated with IR. Excessive glutamate can potentially hinder the insulin signaling pathway through various mechanisms, including the activation of ectopic lipid accumulation, inflammation, and endoplasmic reticulum stress. Glutamate can also modify mitochondrial function through Ca2+ and induce purine degradation mediated by AMP deaminase 2. Exercise has the potential to decrease circulating levels of glutamate, which can be attributed to accelerated glutamate catabolism and enhanced glutamate uptake. SIGNIFICANCE Glutamate may act as a mediator in the exercise-induced improvement of insulin sensitivity.
Collapse
Affiliation(s)
- Xiaorui Xing
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Qin Sun
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Ruwen Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Yibing Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
4
|
Vanni E, Lindner K, Gavin AC, Montessuit C. Differential intracellular management of fatty acids impacts on metabolic stress-stimulated glucose uptake in cardiomyocytes. Sci Rep 2023; 13:14805. [PMID: 37684349 PMCID: PMC10491837 DOI: 10.1038/s41598-023-42072-7] [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: 03/03/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Stimulation of glucose uptake in response to ischemic metabolic stress is important for cardiomyocyte function and survival. Chronic exposure of cardiomyocytes to fatty acids (FA) impairs the stimulation of glucose uptake, whereas induction of lipid droplets (LD) is associated with preserved glucose uptake. However, the mechanisms by which LD induction prevents glucose uptake impairment remain elusive. We induced LD with either tetradecanoyl phorbol acetate (TPA) or 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR). Triacylglycerol biosynthesis enzymes were inhibited in cardiomyocytes exposed to FA ± LD inducers, either upstream (glycerol-3-phosphate acyltransferases; GPAT) or downstream (diacylglycerol acyltransferases; DGAT) of the diacylglycerol step. Although both inhibitions reduced LD formation in cardiomyocytes treated with FA and LD inducers, only DGAT inhibition impaired metabolic stress-stimulated glucose uptake. DGAT inhibition in FA plus TPA-treated cardiomyocytes reduced triacylglycerol but not diacylglycerol content, thus increasing the diacylglycerol/triacylglycerol ratio. In cardiomyocytes exposed to FA alone, GPAT inhibition reduced diacylglycerol but not triacylglycerol, thus decreasing the diacylglycerol/triacylglycerol ratio, prevented PKCδ activation and improved metabolic stress-stimulated glucose uptake. Changes in AMP-activated Protein Kinase activity failed to explain variations in metabolic stress-stimulated glucose uptake. Thus, LD formation regulates metabolic stress-stimulated glucose uptake in a manner best reflected by the diacylglycerol/triacylglycerol ratio.
Collapse
Affiliation(s)
- Ettore Vanni
- Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland
| | - Karina Lindner
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, Geneva, Switzerland
| | - Anne-Claude Gavin
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, Geneva, Switzerland
| | - Christophe Montessuit
- Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland.
| |
Collapse
|
5
|
Morgunova GV, Shilovsky GA, Khokhlov AN. Effect of Caloric Restriction on Aging: Fixing the Problems of Nutrient Sensing in Postmitotic Cells? BIOCHEMISTRY. BIOKHIMIIA 2021; 86:1352-1367. [PMID: 34903158 DOI: 10.1134/s0006297921100151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The review discusses the role of metabolic disorders (in particular, insulin resistance) in the development of age-related diseases and normal aging with special emphasis on the changes in postmitotic cells of higher organisms. Caloric restriction helps to prevent such metabolic disorders, which could probably explain its ability to prolong the lifespan of laboratory animals. Maintaining metabolic homeostasis is especially important for the highly differentiated long-lived body cells, whose lifespan is comparable to the lifespan of the organism itself. Normal functioning of these cells can be ensured only upon correct functioning of the cytoplasm clean-up system and availability of all required nutrients and energy sources. One of the central problems in gerontology is the age-related disruption of glucose metabolism leading to obesity, diabetes, metabolic syndrome, and other related pathologies. Along with the adipose tissue, skeletal muscles are the main consumers of insulin; hence the physical activity of muscles, which supports their energy metabolism, delays the onset of insulin resistance. Insulin resistance disrupts the metabolism of cardiomyocytes, so that they fail to utilize the nutrients to perform their functions even being surrounded by a nutrient-rich environment, which contributes to the development of age-related cardiovascular diseases. Metabolic pathologies also alter the nutrient sensitivity of neurons, thus disrupting the action of insulin in the central nervous system. In addition, there is evidence that neurons can develop insulin resistance as well. It has been suggested that affecting nutritional sensors (e.g., AMPK) in postmitotic cells might improve the state of the entire multicellular organism, slow down its aging, and increase the lifespan.
Collapse
Affiliation(s)
- Galina V Morgunova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Gregory A Shilovsky
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | | |
Collapse
|
6
|
Wang S, Schianchi F, Neumann D, Wong LY, Sun A, van Nieuwenhoven FA, Zeegers MP, Strzelecka A, Col U, Glatz JFC, Nabben M, Luiken JJFP. Specific amino acid supplementation rescues the heart from lipid overload-induced insulin resistance and contractile dysfunction by targeting the endosomal mTOR-v-ATPase axis. Mol Metab 2021; 53:101293. [PMID: 34265467 PMCID: PMC8350375 DOI: 10.1016/j.molmet.2021.101293] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 01/25/2023] Open
Abstract
Objective The diabetic heart is characterized by extensive lipid accumulation which often leads to cardiac contractile dysfunction. The underlying mechanism involves a pivotal role for vacuolar-type H+-ATPase (v-ATPase, functioning as endosomal/lysosomal proton pump). Specifically, lipid oversupply to the heart causes disassembly of v-ATPase and endosomal deacidification. Endosomes are storage compartments for lipid transporter CD36. However, upon endosomal deacidification, CD36 is expelled to translocate to the sarcolemma, thereby inducing myocardial lipid accumulation, insulin resistance, and contractile dysfunction. Hence, the v-ATPase assembly may be a suitable target for ameliorating diabetic cardiomyopathy. Another function of v-ATPase involves the binding of anabolic master-regulator mTORC1 to endosomes, a prerequisite for the activation of mTORC1 by amino acids (AAs). We examined whether the relationship between v-ATPase and mTORC1 also operates reciprocally; specifically, whether AA induces v-ATPase reassembly in a mTORC1-dependent manner to prevent excess lipids from entering and damaging the heart. Methods Lipid overexposed rodent/human cardiomyocytes and high-fat diet-fed rats were treated with a specific cocktail of AAs (lysine/leucine/arginine). Then, v-ATPase assembly status/activity, cell surface CD36 content, myocellular lipid uptake/accumulation, insulin sensitivity, and contractile function were measured. To elucidate underlying mechanisms, specific gene knockdown was employed, followed by subcellular fractionation, and coimmunoprecipitation. Results In lipid-overexposed cardiomyocytes, lysine/leucine/arginine reinternalized CD36 to the endosomes, prevented/reversed lipid accumulation, preserved/restored insulin sensitivity, and contractile function. These beneficial AA actions required the mTORC1–v-ATPase axis, adaptor protein Ragulator, and endosomal/lysosomal AA transporter SLC38A9, indicating an endosome-centric inside-out AA sensing mechanism. In high-fat diet-fed rats, lysine/leucine/arginine had similar beneficial actions at the myocellular level as in vitro in lipid-overexposed cardiomyocytes and partially reversed cardiac hypertrophy. Conclusion Specific AAs acting through v-ATPase reassembly reduce cardiac lipid uptake raising the possibility for treatment in situations of lipid overload and associated insulin resistance. • High physiological concentrations of specific AAs (K/L/R) activate v-ATPase. • The KLR mix activates v-ATPase by mutually dependent activation of mTORC1. • KLR-induced v-ATPase activation enables endosomes to retain lipid transporter CD36. • KLR mends lipid-induced insulin resistance and cardiomyocytic contractile dysfunction. • KLR reverses v-ATPase disassembly and cardiac hypertrophy in high-fat diet-fed rats.
Collapse
Affiliation(s)
- Shujin Wang
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; Institute of Life Sciences, Chongqing Medical University, Chongqing, PR China
| | - Francesco Schianchi
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Dietbert Neumann
- Department of Pathology, Maastricht University Medical Center+, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| | - Li-Yen Wong
- Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Aomin Sun
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Frans A van Nieuwenhoven
- Department of Physiology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| | - Maurice P Zeegers
- Department of Complex Genetics and Epidemiology, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Agnieszka Strzelecka
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Umare Col
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Jan F C Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Miranda Nabben
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| | - Joost J F P Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
7
|
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
|
8
|
Li L, Aslam M, Siegler BH, Niemann B, Rohrbach S. Comparative Analysis of CTRP-Mediated Effects on Cardiomyocyte Glucose Metabolism: Cross Talk between AMPK and Akt Signaling Pathway. Cells 2021; 10:cells10040905. [PMID: 33919975 PMCID: PMC8070942 DOI: 10.3390/cells10040905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/25/2022] Open
Abstract
C1q/tumor necrosis factor -alpha-related proteins (CTRPs) have been shown to mediate protective cardiovascular effects, but no data exists on their effects on glucose and fatty acid (FA) metabolism in cardiomyocytes. In the present study, adult rat cardiomyocytes and H9C2 cardiomyoblasts were stimulated with various recombinant CTRPs. Glucose or FA uptake, expression of genes involved in glucose or FA metabolism and the role of the AMP-activated protein kinase (AMPK) and Akt were investigated. Although most CTRPs induced an increase in phosphorylation of AMPK and Akt in cardiomyocytes, mainly CTRP2, 7, 9 and 13 induced GLUT1 and GLUT4 translocation and glucose uptake in cardiomyocytes, despite high structural similarities among CTRPs. AMPK inhibition reduced the CTRPs-mediated activation of Akt, while Akt inhibition did not impair AMPK activation. In addition, CTRP2, 7, 9 and 13 mediated strong effects on the expression of enzymes involved in glucose or FA metabolism. Loss of adiponectin receptor 1, which has been suggested to be involved in CTRP-induced signal transduction, abolished the effects of some but not all CTRPs on glucose metabolism. Targeting the AMPK signaling pathway via CTRPs may offer a therapeutic principle to restore glucose homeostasis by acting on glucose uptake independent of the Akt pathway.
Collapse
Affiliation(s)
- Ling Li
- Institute of Physiology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.H.S.); (S.R.)
- Correspondence: ; Tel.: +49-641-99-47342
| | - Muhammad Aslam
- Experimental Cardiology, Department of Cardiology and Angiology, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Benedikt H. Siegler
- Institute of Physiology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.H.S.); (S.R.)
| | - Bernd Niemann
- Department of Cardiac and Vascular Surgery, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.H.S.); (S.R.)
| |
Collapse
|
9
|
Ferté L, Marino A, Battault S, Bultot L, Van Steenbergen A, Bol A, Cumps J, Ginion A, Koepsell H, Dumoutier L, Hue L, Horman S, Bertrand L, Beauloye C. New insight in understanding the contribution of SGLT1 in cardiac glucose uptake: evidence for a truncated form in mice and humans. Am J Physiol Heart Circ Physiol 2021; 320:H838-H853. [PMID: 33416451 PMCID: PMC8082801 DOI: 10.1152/ajpheart.00736.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 01/10/2023]
Abstract
Although sodium glucose cotransporter 1 (SGLT1) has been identified as one of the major SGLT isoforms expressed in the heart, its exact role remains elusive. Evidence using phlorizin, the most common inhibitor of SGLTs, has suggested its role in glucose transport. However, phlorizin could also affect classical facilitated diffusion via glucose transporters (GLUTs), bringing into question the relevance of SGLT1 in overall cardiac glucose uptake. Accordingly, we assessed the contribution of SGLT1 in cardiac glucose uptake using the SGLT1 knockout mouse model, which lacks exon 1. Glucose uptake was similar in cardiomyocytes isolated from SGLT1-knockout (Δex1KO) and control littermate (WT) mice either under basal state, insulin, or hyperglycemia. Similarly, in vivo basal and insulin-stimulated cardiac glucose transport measured by micro-PET scan technology did not differ between WT and Δex1KO mice. Micromolar concentrations of phlorizin had no impact on glucose uptake in either isolated WT or Δex1KO-derived cardiomyocytes. However, higher concentrations (1 mM) completely inhibited insulin-stimulated glucose transport without affecting insulin signaling nor GLUT4 translocation independently from cardiomyocyte genotype. Interestingly, we discovered that mouse and human hearts expressed a shorter slc5a1 transcript, leading to SGLT1 protein lacking transmembrane domains and residues involved in glucose and sodium bindings. In conclusion, cardiac SGLT1 does not contribute to overall glucose uptake, probably due to the expression of slc5a1 transcript variant. The inhibitory effect of phlorizin on cardiac glucose uptake is SGLT1-independent and can be explained by GLUT transporter inhibition. These data open new perspectives in understanding the role of SGLT1 in the heart.NEW & NOTEWORTHY Ever since the discovery of its expression in the heart, SGLT1 has been considered as similar as the intestine and a potential contributor to cardiac glucose transport. For the first time, we have demonstrated that a slc5a1 transcript variant is present in the heart that has no significant impact on cardiac glucose handling.
Collapse
Affiliation(s)
- Laura Ferté
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Alice Marino
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Sylvain Battault
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Laurent Bultot
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Anne Van Steenbergen
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Anne Bol
- Center of Molecular Imaging, Radiotherapy and Oncology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Julien Cumps
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Audrey Ginion
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Hermann Koepsell
- Department of Molecular Plant Physiology and Biophysics, Julius von Sachs Institute, University of Würzburg, Würzburg, Germany
| | - Laure Dumoutier
- Médecine Expérimentale, Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
| | - Louis Hue
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
- Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
| | - Sandrine Horman
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Luc Bertrand
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Christophe Beauloye
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
10
|
Haye A, Ansari MA, Rahman SO, Shamsi Y, Ahmed D, Sharma M. Role of AMP-activated protein kinase on cardio-metabolic abnormalities in the development of diabetic cardiomyopathy: A molecular landscape. Eur J Pharmacol 2020; 888:173376. [PMID: 32810493 DOI: 10.1016/j.ejphar.2020.173376] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
Cardiovascular complications associated with diabetes mellitus remains a leading cause of morbidity and mortality across the world. Diabetic cardiomyopathy is a descriptive pathology that in absence of co-morbidities such as hypertension, dyslipidemia initially characterized by cardiac stiffness, myocardial fibrosis, ventricular hypertrophy, and remodeling. These abnormalities further contribute to diastolic dysfunctions followed by systolic dysfunctions and eventually results in clinical heart failure (HF). The clinical outcomes associated with HF are considerably worse in patients with diabetes. The complexity of the pathogenesis and clinical features of diabetic cardiomyopathy raises serious questions in developing a therapeutic strategy to manage cardio-metabolic abnormalities. Despite extensive research in the past decade the compelling approaches to manage and treat diabetic cardiomyopathy are limited. AMP-Activated Protein Kinase (AMPK), a serine-threonine kinase, often referred to as cellular "metabolic master switch". During the development and progression of diabetic cardiomyopathy, a plethora of evidence demonstrate the beneficial role of AMPK on cardio-metabolic abnormalities including altered substrate utilization, impaired cardiac insulin metabolic signaling, mitochondrial dysfunction and oxidative stress, myocardial inflammation, increased accumulation of advanced glycation end-products, impaired cardiac calcium handling, maladaptive activation of the renin-angiotensin-aldosterone system, endoplasmic reticulum stress, myocardial fibrosis, ventricular hypertrophy, cardiac apoptosis, and impaired autophagy. Therefore, in this review, we have summarized the findings from pre-clinical and clinical studies and provided a collective overview of the pathophysiological mechanism and the regulatory role of AMPK on cardio-metabolic abnormalities during the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Abdul Haye
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd Asif Ansari
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Syed Obaidur Rahman
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Yasmeen Shamsi
- Department of Moalejat, School of Unani Medical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Danish Ahmed
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom University of Agriculture Technology and Sciences, Allahabad, Uttar Pradesh, India
| | - Manju Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
11
|
Battault S, Renguet E, Van Steenbergen A, Horman S, Beauloye C, Bertrand L. Myocardial glucotoxicity: Mechanisms and potential therapeutic targets. Arch Cardiovasc Dis 2020; 113:736-748. [PMID: 33189592 DOI: 10.1016/j.acvd.2020.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022]
Abstract
Besides coronary artery disease, which remains the main cause of heart failure in patients with diabetes, factors independent of coronary artery disease are involved in the development of heart failure in the onset of what is called diabetic cardiomyopathy. Among them, hyperglycaemia - a hallmark of type 2 diabetes - has both acute and chronic deleterious effects on myocardial function, and clearly participates in the establishment of diabetic cardiomyopathy. In the present review, we summarize the cellular and tissular events that occur in a heart exposed to hyperglycaemia, and depict the complex molecular mechanisms proposed to be involved in glucotoxicity. Finally, from a more translational perspective, different therapeutic strategies targeting hyperglycaemia-mediated molecular mechanisms will be detailed.
Collapse
Affiliation(s)
- Sylvain Battault
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Edith Renguet
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Anne Van Steenbergen
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Sandrine Horman
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Christophe Beauloye
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium; Division of cardiology, Cliniques Universitaires Saint-Luc, B-1200 Brussels, Belgium.
| | - Luc Bertrand
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium; WELBIO, B-1300 Wavre, Belgium.
| |
Collapse
|
12
|
Bertrand L, Auquier J, Renguet E, Angé M, Cumps J, Horman S, Beauloye C. Glucose transporters in cardiovascular system in health and disease. Pflugers Arch 2020; 472:1385-1399. [PMID: 32809061 DOI: 10.1007/s00424-020-02444-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022]
Abstract
Glucose transporters are essential for the heart to sustain its function. Due to its nature as a high energy-consuming organ, the heart needs to catabolize a huge quantity of metabolic substrates. For optimized energy production, the healthy heart constantly switches between various metabolites in accordance with substrate availability and hormonal status. This metabolic flexibility is essential for the maintenance of cardiac function. Glucose is part of the main substrates catabolized by the heart and its use is fine-tuned via complex molecular mechanisms that include the regulation of the glucose transporters GLUTs, mainly GLUT4 and GLUT1. Besides GLUTs, glucose can also be transported by cotransporters of the sodium-glucose cotransporter (SGLT) (SLC5 gene) family, in which SGLT1 and SMIT1 were shown to be expressed in the heart. This SGLT-mediated uptake does not seem to be directly linked to energy production but is rather associated with intracellular signalling triggering important processes such as the production of reactive oxygen species. Glucose transport is markedly affected in cardiac diseases such as cardiac hypertrophy, diabetic cardiomyopathy and heart failure. These alterations are not only fingerprints of these diseases but are involved in their onset and progression. The present review will depict the importance of glucose transport in healthy and diseased heart, as well as proposed therapies targeting glucose transporters.
Collapse
Affiliation(s)
- Luc Bertrand
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium.
| | - Julien Auquier
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium
| | - Edith Renguet
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium
| | - Marine Angé
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium
| | - Julien Cumps
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium
| | - Sandrine Horman
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium
| | - Christophe Beauloye
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium.,Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
13
|
Chronic AICAR treatment prevents metabolic changes in cardiomyocytes exposed to free fatty acids. Pflugers Arch 2019; 471:1219-1234. [PMID: 31152240 DOI: 10.1007/s00424-019-02285-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/27/2019] [Accepted: 05/15/2019] [Indexed: 01/09/2023]
Abstract
The stimulation of glucose transport by metabolic stress is an important determinant of myocardial susceptibility to ischemia and reperfusion injury. Stimulation of glucose transport is markedly impaired in cardiomyocytes chronically exposed to excess free fatty acids (FFA), as occurs in vivo in type 2 diabetes. To determine whether chronic low-grade activation of AMP-activated kinase (AMPK) improves substrate metabolism in cardiomyocytes exposed to FFA, isolated cultured cardiomyocytes were exposed for 7 days to FFA ± the AMPK agonist 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR). Glucose transport and glycolysis were then measured during acute metabolic stress provoked by oligomycin. Chronic treatment with AICAR improved basal and oligomycin-stimulated glucose transport in FFA-exposed but not in control cardiomyocytes. Similarly, basal and oligomycin-stimulated glycolysis was reduced in FFA-exposed cardiomyocytes but restored by chronic AICAR treatment. Conversely, fatty acid oxidation was increased in FFA-exposed cardiomyocytes and reduced by chronic AICAR treatment. Chronic AICAR treatment induced in FFA-exposed cardiomyocytes the biogenesis of numerous lipid droplets. Curiously, whereas acute treatment of cardiomyocytes with AICAR increased phosphorylation of the AMPKα subunit on T172, a classical marker of AMPK activation, chronic AICAR treatment almost completely obliterated T172 phosphorylation. However, phosphorylation of the AMPK target protein raptor on S792 was reduced in FFA-exposed cardiomyocytes but restored by AICAR treatment. In conclusion, chronic AICAR treatment induces a metabolic shift in FFA-exposed cardiomyocytes, characterized by improved glucose transport and glycolysis and redirection of fatty acids towards neutral storage. Such metabolic changes in vivo could protect the hearts of patients with type 2 diabetes against ischemia-reperfusion injury.
Collapse
|
14
|
Aragón-Herrera A, Feijóo-Bandín S, Rodríguez-Penas D, Roselló-Lletí E, Portolés M, Rivera M, Bigazzi M, Bani D, Gualillo O, González-Juanatey JR, Lago F. Relaxin activates AMPK-AKT signaling and increases glucose uptake by cultured cardiomyocytes. Endocrine 2018; 60:103-111. [PMID: 29411306 DOI: 10.1007/s12020-018-1534-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 01/15/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE Many evidences show that the hormone relaxin plays a pivotal role in the physiology and pathology of the cardiovascular system. This pleiotropic hormone exerts regulatory functions through specific receptors in cardiovascular tissues: in experimental animal models it was shown to induce coronary vasodilation, prevent cardiac damage induced by ischemia/reperfusion and revert cardiac hypertrophy and fibrosis. A tight relationship between this hormone and important metabolic pathways has been suggested, but it is at present unknown if relaxin could regulate cardiac metabolism. Our aim was to study the possible effects of relaxin on cardiomyocyte metabolism. METHODS Neonatal rat cardiomyocytes were treated with relaxin and (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assays (MTT) were performed to assess metabolic activity; while 2-deoxy-D-[3H] glucose and BODIPY-labelled fatty acid incorporations were analyzed to measure glucose and fatty acid uptakes, and western blot was utilized to study the intracellular signaling pathways activated by the hormone. RESULTS We observed that relaxin at 10 ng/ml was able to increase the level of metabolic activity of cultured neonatal rat cardiomyocytes; the rate of 2-deoxy-D-[3H]glucose incorporation demonstrated that relaxin also induced an increase in glucose uptake. First evidence is also offered that relaxin can activate the master energy sensor and regulator AMPK in cardiomyocytes. Moreover, the treatment of cardiomyocytes with relaxin also induced dose-dependent increases in ERK1/2, AKT, and AS160 phosphorylation. That raise in AS160 phosphorylation induced by relaxin was prevented by the pretreatment with AMPK and AKT pathways inhibitors, indicating that both molecules play important roles in the relaxin effects reported. CONCLUSION Relaxin can regulate cardiomyocyte metabolism and activate AMPK, the central sensor of energy status that maintains cellular energy homeostasis, and also ERK and AKT, two molecular sensing nodes that coordinate dynamic responses of the cell's metabolic responses.
Collapse
Affiliation(s)
- A Aragón-Herrera
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - S Feijóo-Bandín
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain.
- CIBERCV, Institute of Health Carlos III, Madrid, Spain.
| | - D Rodríguez-Penas
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - E Roselló-Lletí
- CIBERCV, Institute of Health Carlos III, Madrid, Spain
- Cardiocirculatory Unit, Health Institute of La Fe University Hospital (IIS La Fe), Valencia, Spain
| | - M Portolés
- CIBERCV, Institute of Health Carlos III, Madrid, Spain
- Cardiocirculatory Unit, Health Institute of La Fe University Hospital (IIS La Fe), Valencia, Spain
| | - M Rivera
- CIBERCV, Institute of Health Carlos III, Madrid, Spain
- Cardiocirculatory Unit, Health Institute of La Fe University Hospital (IIS La Fe), Valencia, Spain
| | - M Bigazzi
- Prosperius Institute, Florence, Italy
| | - D Bani
- Prosperius Institute, Florence, Italy
| | - O Gualillo
- Neuroendocrine Interaccions in Rheumatology and Inflammatory Diseases Unit, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - J R González-Juanatey
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
- CIBERCV, Institute of Health Carlos III, Madrid, Spain
| | - F Lago
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
- CIBERCV, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
15
|
Fang X, Shen F, Lechauve C, Xu P, Zhao G, Itkow J, Wu F, Hou Y, Wu X, Yu L, Xiu H, Wang M, Zhang R, Wang F, Zhang Y, Wang D, Weiss MJ, Yu D. miR-144/451 represses the LKB1/AMPK/mTOR pathway to promote red cell precursor survival during recovery from acute anemia. Haematologica 2017; 103:406-416. [PMID: 29269522 PMCID: PMC5830375 DOI: 10.3324/haematol.2017.177394] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/20/2017] [Indexed: 12/21/2022] Open
Abstract
The microRNAs miR-144 and -451 are encoded by a bicistronic gene that is strongly induced during red blood cell formation (erythropoiesis). Ablation of the miR-144/451 gene in mice causes mild anemia under baseline conditions. Here we show that miR-144/451−/− erythroblasts exhibit increased apoptosis during recovery from acute anemia. Mechanistically, miR-144/451 depletion increases the expression of the miR-451 target mRNA Cab39, which encodes a co-factor for the serine-threonine kinase LKB1. During erythropoietic stress, miR-144/451−/− erythroblasts exhibit abnormally increased Cab39 protein, which activates LKB1 and its downstream AMPK/mTOR effector pathway. Suppression of this pathway via drugs or shRNAs enhances survival of the mutant erythroblasts. Thus, miR-144/451 facilitates recovery from acute anemia by repressing Cab39/AMPK/mTOR. Our findings suggest that miR-144/451 is a key protector of erythroblasts during pathological states associated with dramatically increased erythropoietic demand, including acute blood loss and hemolytic anemia.
Collapse
Affiliation(s)
- Xiao Fang
- Clinical Medical College of Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China
| | - Feiyang Shen
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China
| | - Christophe Lechauve
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peng Xu
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Guowei Zhao
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jacobi Itkow
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China
| | - Fan Wu
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China
| | - Yaying Hou
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China
| | - Xiaohui Wu
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China.,Department of Pediatrics, Jingjiang People's Hospital, Yangzhou University, Jingjiang, China
| | - Lingling Yu
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China.,Department of Pediatrics, Jingjiang People's Hospital, Yangzhou University, Jingjiang, China
| | - Huiqing Xiu
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China
| | - Mengli Wang
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China
| | - Ruiling Zhang
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China
| | - Fangfang Wang
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China
| | - Yanqing Zhang
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China
| | - Daxin Wang
- Clinical Medical College of Yangzhou University, Yangzhou, China
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Duonan Yu
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, University School of Medicine, China .,Institute of Comparative Medicine, Yangzhou University, China.,Institute of Translational Medicine, Yangzhou University School of Medicine, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou, China
| |
Collapse
|
16
|
Renguet E, Ginion A, Gélinas R, Bultot L, Auquier J, Robillard Frayne I, Daneault C, Vanoverschelde JL, Des Rosiers C, Hue L, Horman S, Beauloye C, Bertrand L. Metabolism and acetylation contribute to leucine-mediated inhibition of cardiac glucose uptake. Am J Physiol Heart Circ Physiol 2017. [PMID: 28646031 DOI: 10.1152/ajpheart.00738.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
High plasma leucine levels strongly correlate with type 2 diabetes. Studies of muscle cells have suggested that leucine alters the insulin response for glucose transport by activating an insulin-negative feedback loop driven by the mammalian target of rapamycin/p70 ribosomal S6 kinase (mTOR/p70S6K) pathway. Here, we examined the molecular mechanism involved in leucine's action on cardiac glucose uptake. Leucine was indeed able to curb glucose uptake after insulin stimulation in both cultured cardiomyocytes and perfused hearts. Although leucine activated mTOR/p70S6K, the mTOR inhibitor rapamycin did not prevent leucine's inhibitory action on glucose uptake, ruling out the contribution of the insulin-negative feedback loop. α-Ketoisocaproate, the first metabolite of leucine catabolism, mimicked leucine's effect on glucose uptake. Incubation of cardiomyocytes with [13C]leucine ascertained its metabolism to ketone bodies (KBs), which had a similar negative impact on insulin-stimulated glucose transport. Both leucine and KBs reduced glucose uptake by affecting translocation of glucose transporter 4 (GLUT4) to the plasma membrane. Finally, we found that leucine elevated the global protein acetylation level. Pharmacological inhibition of lysine acetyltransferases counteracted this increase in protein acetylation and prevented leucine's inhibitory action on both glucose uptake and GLUT4 translocation. Taken together, these results indicate that leucine metabolism into KBs contributes to inhibition of cardiac glucose uptake by hampering the translocation of GLUT4-containing vesicles via acetylation. They offer new insights into the establishment of insulin resistance in the heart.NEW & NOTEWORTHY Catabolism of the branched-chain amino acid leucine into ketone bodies efficiently inhibits cardiac glucose uptake through decreased translocation of glucose transporter 4 to the plasma membrane. Leucine increases protein acetylation. Pharmacological inhibition of acetylation reverses leucine's action, suggesting acetylation involvement in this phenomenon.Listen to this article's corresponding podcast at http://ajpheart.podbean.com/e/leucine-metabolism-inhibits-cardiac-glucose-uptake/.
Collapse
Affiliation(s)
- Edith Renguet
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Audrey Ginion
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Roselle Gélinas
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Laurent Bultot
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Julien Auquier
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | | | | | - Jean-Louis Vanoverschelde
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium.,Cliniques Universitaires Saint-Luc, Division of Cardiology, Brussels, Belgium
| | - Christine Des Rosiers
- Montreal Heart Institute, Montreal, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; and
| | - Louis Hue
- Université catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Sandrine Horman
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Christophe Beauloye
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium.,Cliniques Universitaires Saint-Luc, Division of Cardiology, Brussels, Belgium
| | - Luc Bertrand
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium;
| |
Collapse
|
17
|
Freitag H, Christen F, Lewens F, Grass I, Briest F, Iwaszkiewicz S, Siegmund B, Grabowski P. Inhibition of mTOR's Catalytic Site by PKI-587 Is a Promising Therapeutic Option for Gastroenteropancreatic Neuroendocrine Tumor Disease. Neuroendocrinology 2017; 105:90-104. [PMID: 27513674 PMCID: PMC5475233 DOI: 10.1159/000448843] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 08/02/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND The characteristic clinical heterogeneity and mostly slow-growing behavior of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) cause problems in finding appropriate treatments. Thus, the current therapy options are not satisfactory. PKI-587 is a highly potent, novel dual inhibitor of PI3K and mTORC1/C2. AIM We assessed the effects of PKI-587 in different GEP-NEN tumor models, including the poorly differentiated cell line LCC-18, and compared them with those of the established mTORC1 inhibitor everolimus. METHODS We treated BON, QGP-1, KRJ-I, and LCC-18 cell lines with increasing concentrations of the inhibitor PKI-587, and compared the results with those of everolimus and DMSO. We assessed the impact of the treatments on viability (WST-1 assay), on apoptotic processes (caspase 3/7 assay, JC-1), and on cell cycle regulation (flow cytometry). We determined alterations in signaling mediators by phosphor-specific Western blot analysis and conducted multiplexed gene expression analysis (nCounter® technology). RESULTS In all cell lines, PKI-587 dose-dependently inhibited proliferation, whereas everolimus was less effective. Treatment with PKI-587 led to cell cycle arrest and induction of apoptosis and successfully suppressed activity of the direct mTORC1 target 4E-BP1, a crucial factor for tumor genesis only partially inhibited by everolimus. Gene expression analyses revealed relevant changes of RAS, MAPK, STAT, and PI3K pathway genes after treatment. Treatment-dependent and cell line-characteristic effects on AKT/Rb/E2F signaling regarding cell cycle control and apoptosis are extensively discussed in this paper. CONCLUSION PI3K/mTOR dual targeting is a promising new therapeutic approach in neuroendocrine tumor disease that should be evaluated in further clinical trials.
Collapse
Affiliation(s)
- Helma Freitag
- Department of Gastroenterology, Infectious Diseases, Rheumatology CC13, Charité - Universitätsmedizin Berlin, Germany
| | - Friederike Christen
- Department of Gastroenterology, Infectious Diseases, Rheumatology CC13, Charité - Universitätsmedizin Berlin, Germany
- Institute of Biology, Humboldt-Universität Berlin, Berlin, Germany
| | - Florentine Lewens
- Department of Gastroenterology, Infectious Diseases, Rheumatology CC13, Charité - Universitätsmedizin Berlin, Germany
| | - Irina Grass
- Department of Gastroenterology, Infectious Diseases, Rheumatology CC13, Charité - Universitätsmedizin Berlin, Germany
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Franziska Briest
- Department of Gastroenterology, Infectious Diseases, Rheumatology CC13, Charité - Universitätsmedizin Berlin, Germany
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Sara Iwaszkiewicz
- Department of Gastroenterology, Infectious Diseases, Rheumatology CC13, Charité - Universitätsmedizin Berlin, Germany
- Institute of Biology, Humboldt-Universität Berlin, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases, Rheumatology CC13, Charité - Universitätsmedizin Berlin, Germany
| | - Patricia Grabowski
- Department of Gastroenterology, Infectious Diseases, Rheumatology CC13, Charité - Universitätsmedizin Berlin, Germany
- Department of Gastroenterology and Endocrinology, Zentralklinik Bad Berka GmbH, Bad Berka, Germany
- *Patricia Grabowski, Department of Gastroenterology, Infectious Diseases, Rheumatology CC13, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, DE-12203 Berlin (Germany), E-Mail
| |
Collapse
|
18
|
Trivedi PC, Bartlett JJ, Perez LJ, Brunt KR, Legare JF, Hassan A, Kienesberger PC, Pulinilkunnil T. Glucolipotoxicity diminishes cardiomyocyte TFEB and inhibits lysosomal autophagy during obesity and diabetes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1893-1910. [PMID: 27620487 DOI: 10.1016/j.bbalip.2016.09.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 01/07/2023]
Abstract
Impaired cardiac metabolism in the obese and diabetic heart leads to glucolipotoxicity and ensuing cardiomyopathy. Glucolipotoxicity causes cardiomyocyte injury by increasing energy insufficiency, impairing proteasomal-mediated protein degradation and inducing apoptosis. Proteasome-evading proteins are degraded by autophagy in the lysosome, whose metabolism and function are regulated by master regulator transcription factor EB (TFEB). Limited studies have examined the impact of glucolipotoxicity on intra-lysosomal signaling proteins and their regulators. By utilizing a mouse model of diet-induced obesity, type-1 diabetes (Akita) and ex-vivo model of glucolipotoxicity (H9C2 cells and NRCM, neonatal rat cardiomyocyte), we examined whether glucolipotoxicity negatively targets TFEB and lysosomal proteins to dysregulate autophagy and cause cardiac injury. Despite differential effects of obesity and diabetes on LC3B-II, expression of proteins facilitating autophagosomal clearance such as TFEB, LAMP-2A, Hsc70 and Hsp90 were decreased in the obese and diabetic heart. In-vivo data was recapitulated in H9C2 and NRCM cells, which exhibited impaired autophagic flux and reduced TFEB content when exposed to a glucolipotoxic milieu. Notably, overloading myocytes with a saturated fatty acid (palmitate) but not an unsaturated fatty acid (oleate) depleted cellular TFEB and suppressed autophagy, suggesting a fatty acid specific regulation of TFEB and autophagy in the cardiomyocyte. The effect of glucolipotoxicity to reduce TFEB content was also confirmed in heart tissue from patients with Class-I obesity. Therefore, during glucolipotoxicity, suppression of lysosomal autophagy was associated with reduced lysosomal content, decreased cathepsin-B activity and diminished cellular TFEB content likely rendering myocytes susceptible to cardiac injury.
Collapse
Affiliation(s)
- Purvi C Trivedi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John E2L4L5, New Brunswick, Canada
| | - Jordan J Bartlett
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John E2L4L5, New Brunswick, Canada
| | - Lester J Perez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John E2L4L5, New Brunswick, Canada
| | - Keith R Brunt
- Deparment of Pharmacology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John E2L4L5, New Brunswick, Canada
| | - Jean Francois Legare
- Department of Surgery, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John E2L4L5, New Brunswick, Canada
| | - Ansar Hassan
- Department of Surgery, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John E2L4L5, New Brunswick, Canada
| | - Petra C Kienesberger
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John E2L4L5, New Brunswick, Canada
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John E2L4L5, New Brunswick, Canada.
| |
Collapse
|
19
|
Chan YK, Sung HK, Jahng JWS, Kim GHE, Han M, Sweeney G. Lipocalin-2 inhibits autophagy and induces insulin resistance in H9c2 cells. Mol Cell Endocrinol 2016; 430:68-76. [PMID: 27090568 DOI: 10.1016/j.mce.2016.04.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/05/2016] [Accepted: 04/13/2016] [Indexed: 01/08/2023]
Abstract
Lipocalin-2 (Lcn2; also known as neutrophil gelatinase associated lipocalin, NGAL) levels are increased in obesity and diabetes and associate with insulin resistance. Correlations exist between Lcn2 levels and various forms or stages of heart failure. Insulin resistance and autophagy both play well-established roles in cardiomyopathy. However, little is known about the impact of Lcn2 on insulin signaling in cardiomyocytes. In this study, we treated H9c2 cells with recombinant Lcn2 for 1 h followed by dose- and time-dependent insulin treatment and found that Lcn2 attenuated insulin signaling assessed via phosphorylation of Akt and p70S6K. We used multiple assays to demonstrate that Lcn2 reduced autophagic flux. First, Lcn2 reduced pULK1 S555, increased pULK1 S757 and reduced LC3-II levels determined by Western blotting. We validated the use of DQ-BSA to assess autolysosomal protein degradation and this together with MagicRed cathepsin B assay indicated that Lcn2 reduced lysosomal degradative activity. Furthermore, we generated H9c2 cells stably expressing tandem fluorescent RFP/GFP-LC3 and this approach verified that Lcn2 decreased autophagic flux. We also created an autophagy-deficient H9c2 cell model by overexpressing a dominant-negative Atg5 mutant and found that reduced autophagy levels also induced insulin resistance. Adding rapamycin after Lcn2 could stimulate autophagy and recover insulin sensitivity. In conclusion, our study indicated that acute Lcn2 treatment caused insulin resistance and use of gain and loss of function approaches elucidated a causative link between autophagy inhibition and regulation of insulin sensitivity by Lcn2.
Collapse
Affiliation(s)
- Yee Kwan Chan
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | - Hye Kyoung Sung
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | | | - Grace Ha Eun Kim
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | - Meng Han
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
20
|
Bu L, Chang X, Cheng X, Yao Q, Su B, Sheng C, Qu S. Activated central galanin type 1 receptor alleviated insulin resistance in diabetic rat muscle. J Neurosci Res 2016; 94:947-55. [PMID: 27410235 DOI: 10.1002/jnr.23775] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 04/27/2016] [Accepted: 05/02/2016] [Indexed: 01/14/2023]
Affiliation(s)
- Le Bu
- Department of Endocrinology; Shanghai 10th People's Hospital, Tongji University School of Medicine; Shanghai People's Republic of China
| | - Xusheng Chang
- Department of General Surgery; Yancheng City First People's Hospital; Yancheng City Jiangsu People's Republic of China
| | - Xiaoyun Cheng
- Department of Endocrinology; Shanghai 10th People's Hospital, Tongji University School of Medicine; Shanghai People's Republic of China
| | - Qian Yao
- Key Laboratory of Sichuan Province of Medicinal Chemistry; Chengdu University; Chengdu People's Republic of China
| | - Bin Su
- Department of Endocrinology; Shanghai 10th People's Hospital, Tongji University School of Medicine; Shanghai People's Republic of China
| | - Chunjun Sheng
- Department of Endocrinology; Shanghai 10th People's Hospital, Tongji University School of Medicine; Shanghai People's Republic of China
| | - Shen Qu
- Department of Endocrinology; Shanghai 10th People's Hospital, Tongji University School of Medicine; Shanghai People's Republic of China
| |
Collapse
|
21
|
Craps J, Joris V, De Jongh B, Sonveaux P, Horman S, Lengelé B, Bertrand L, Many MC, Colin IM, Gérard AC. Involvement of mTOR and Regulation by AMPK in Early Iodine Deficiency-Induced Thyroid Microvascular Activation. Endocrinology 2016; 157:2545-59. [PMID: 27035650 DOI: 10.1210/en.2015-1911] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Iodine deficiency (ID) induces TSH-independent microvascular activation in the thyroid via the reactive oxygen species/nitric oxide-hypoxia-inducible factor-1α/vascular endothelial growth factor (VEGF) pathway. We hypothesized the additional involvement of mammalian target of rapamycin (mTOR) as a positive regulator of this pathway and AMP-activated protein kinase (AMPK) as a negative feedback regulator to explain the transient nature of ID-induced microvascular changes under nonmalignant conditions. mTOR and AMPK involvement was investigated using an in vitro model (human thyrocytes in primary cultures) and 2 murine models of goitrogenesis (normal NMRI and RET-PTC mice [a papillary thyroid cancer model]). In NMRI mice, ID had no effect on the phosphorylation of ribosomal S6 kinase (p70S6K), a downstream target of mTOR. However, rapamycin inhibited ID-induced thyroid blood flow and VEGF protein expression. In the RET-PTC model, ID strongly increased the phosphorylation of p70S6K, whereas rapamycin completely inhibited the ID-induced increase in p70S6K phosphorylation, thyroid blood flow, and VEGF-A expression. In vitro, although ID increased p70S6K phosphorylation, the ID-stimulated hypoxia-inducible factor/VEGF pathway was inhibited by rapamycin. Activation of AMPK by metformin inhibited ID effects both in vivo and in vitro. In AMPK-α1 knockout mice, the ID-induced increase in thyroid blood flow and VEGF-A protein expression persisted throughout the treatment, whereas both parameters returned to control values in wild-type mice after 4 days of ID. In conclusion, mTOR is required for early ID-induced thyroid microvascular activation. AMPK negatively regulates this pathway, which may account for the transient nature of ID-induced TSH-independent vascular effects under benign conditions.
Collapse
Affiliation(s)
- J Craps
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - V Joris
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - B De Jongh
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - P Sonveaux
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - S Horman
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - B Lengelé
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - L Bertrand
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - M-C Many
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - I M Colin
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| | - A-C Gérard
- Pôle de Morphologie (J.C., B.D.J., B.L., M.-C.M., I.M.C., A.-C.G.), de Pharmacologie et Thérapeutique (V.J., P.S.), de Recherche Cardiovasculaire (S.H., L.B.), Institut de Recherche Expérimentale et Clinique, Laboratoire de Biologie Cellulaire, and Institut des Sciences de la Vie (A.-C.G.), Université Catholique de Louvain, Louvain-La-Neuve, Brussels 1200, Belgium
| |
Collapse
|
22
|
Talagavadi V, Rapisarda P, Galvano F, Pelicci P, Giorgio M. Cyanidin-3- O -β-glucoside and protocatechuic acid activate AMPK/mTOR/S6K pathway and improve glucose homeostasis in mice. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.12.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
23
|
Daskalopoulos EP, Dufeys C, Beauloye C, Bertrand L, Horman S. AMPK in Cardiovascular Diseases. EXPERIENTIA SUPPLEMENTUM (2012) 2016; 107:179-201. [PMID: 27812981 DOI: 10.1007/978-3-319-43589-3_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This chapter summarizes the implication of AMP-activated protein kinase (AMPK) in the regulation of various physiological and pathological cellular events of great importance for the maintenance of cardiac function. These include the control of both metabolic and non-metabolic elements targeting the different cellular components of the cardiac tissue, i.e., cardiomyocytes, fibroblasts, and vascular cells. The description of the multifaceted action of the two AMPK catalytic isoforms, α1 and α2, emphasizes the general protective action of this protein kinase against the development of critical pathologies like myocardial ischemia, cardiac hypertrophy, diabetic cardiomyopathy, and heart failure.
Collapse
Affiliation(s)
- Evangelos P Daskalopoulos
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium.,Cardiovascular Research (Care) Institute, Athens, Ioannina, Greece
| | - Cécile Dufeys
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Christophe Beauloye
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium.,Division of Cardiology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Luc Bertrand
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium.
| | - Sandrine Horman
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| |
Collapse
|
24
|
Regulation of Carbohydrate Metabolism, Lipid Metabolism, and Protein Metabolism by AMPK. EXPERIENTIA SUPPLEMENTUM (2012) 2016; 107:23-43. [PMID: 27812975 DOI: 10.1007/978-3-319-43589-3_2] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This chapter summarizes AMPK function in the regulation of substrate and energy metabolism with the main emphasis on carbohydrate and lipid metabolism, protein turnover, mitochondrial biogenesis, and whole-body energy homeostasis. AMPK acts as whole-body energy sensor and integrates different signaling pathway to meet both cellular and body energy requirements while inhibiting energy-consuming processes but also activating energy-producing ones. AMPK mainly promotes glucose and fatty acid catabolism, whereas it prevents protein, glycogen, and fatty acid synthesis.
Collapse
|
25
|
Franekova V, Angin Y, Hoebers NTH, Coumans WA, Simons PJ, Glatz JFC, Luiken JJFP, Larsen TS. Marine omega-3 fatty acids prevent myocardial insulin resistance and metabolic remodeling as induced experimentally by high insulin exposure. Am J Physiol Cell Physiol 2015; 308:C297-307. [DOI: 10.1152/ajpcell.00073.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Insulin resistance is an important risk factor for the development of several cardiac pathologies, thus advocating strategies for restoring insulin sensitivity of the heart in these conditions. Omega-3 polyunsaturated fatty acids (ω-3 PUFAs), mainly eicosapentaenoic acid (EPA, C20:5n-3) and docosahexaenoic acid (DHA, C22:6n-3), have been shown to improve insulin sensitivity in insulin-sensitive tissues, but their direct effect on insulin signaling and metabolic parameters in the myocardium has not been reported previously. The aim of this study was therefore to examine the ability of EPA and DHA to prevent insulin resistance in isolated rat cardiomyocytes. Primary rat cardiomyocytes were made insulin resistant by 48 h incubation in high insulin (HI) medium. Parallel incubations were supplemented by 200 μM EPA or DHA. Addition of EPA or DHA to the medium prevented the induction of insulin resistance in cardiomyocytes by preserving the phosphorylation state of key proteins in the insulin signaling cascade and by preventing persistent relocation of fatty acid transporter CD36 to the sarcolemma. Only cardiomyocytes incubated in the presence of EPA, however, exhibited improvements in glucose and fatty acid uptake and cell shortening. We conclude that ω-3 PUFAs protect metabolic and functional properties of cardiomyocytes subjected to insulin resistance-evoking conditions.
Collapse
Affiliation(s)
- Veronika Franekova
- Cardiovascular Research Group, Department of Medical Biology, Health Sciences Faculty, UiT The Arctic University of Norway, Tromsø, Norway
| | - Yeliz Angin
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; and
| | - Nicole T. H. Hoebers
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; and
| | - Will A. Coumans
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; and
| | | | - Jan F. C. Glatz
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; and
| | - Joost J. F. P. Luiken
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; and
| | - Terje S. Larsen
- Cardiovascular Research Group, Department of Medical Biology, Health Sciences Faculty, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
26
|
Zhou J, Wang Q, Ding Y, Zou MH. Hypochlorous acid via peroxynitrite activates protein kinase Cθ and insulin resistance in adipocytes. J Mol Endocrinol 2015; 54:25-37. [PMID: 25381390 PMCID: PMC4261204 DOI: 10.1530/jme-14-0213] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We recently reported that genetic deletion of myeloperoxidase (MPO) alleviates obesity-related insulin resistance in mice in vivo. How MPO impairs insulin sensitivity in adipocytes is poorly characterized. As hypochlorous acid (HOCl) is a principal oxidant product generated by MPO, we evaluated the effects of HOCl on insulin signaling in adipocytes differentiated from 3T3-L1 cells. Exposure of 3T3-L1 adipocytes to exogenous HOCl (200 μmol/l) attenuated insulin-stimulated 2-deoxyglucose uptake, GLUT4 translocation, and insulin signals, including tyrosine phosphorylation of insulin receptor substrate 1 (IRS1) and phosphorylation of Akt. Furthermore, treatment with HOCl induced phosphorylation of IRS1 at serine 307, inhibitor κB kinase (IKK), c-Jun NH2-terminal kinase (JNK), and phosphorylation of PKCθ (PKCθ). In addition, genetic and pharmacological inhibition of IKK and JNK abolished serine phosphorylation of IRS1 and impairment of insulin signaling by HOCl. Furthermore, knockdown of PKCθ using siRNA transfection suppressed phosphorylation of IKK and JNK and consequently attenuated the HOCl-impaired insulin signaling pathway. Moreover, activation of PKCθ by peroxynitrite was accompanied by increased phosphorylation of IKK, JNK, and IRS1-serine 307. In contrast, ONOO(-) inhibitors abolished HOCl-induced phosphorylation of PKCθ, IKK, JNK, and IRS1-serine 307, as well as insulin resistance. Finally, high-fat diet (HFD)-induced insulin resistance was associated with enhanced phosphorylation of PKCθ, IKK, JNK, and IRS1 at serine 307 in white adipose tissues from WT mice, all of which were not found in Mpo knockout mice fed HFDs. We conclude that HOCl impairs insulin signaling pathway by increasing ONOO(-) mediated phosphorylation of PKCθ, resulting in phosphorylation of IKK/JNK and consequent serine phosphorylation of IRS1 in adipocytes.
Collapse
Affiliation(s)
- Jun Zhou
- Section of Molecular MedicineBSEB 306A, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Qilong Wang
- Section of Molecular MedicineBSEB 306A, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Ye Ding
- Section of Molecular MedicineBSEB 306A, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Ming-Hui Zou
- Section of Molecular MedicineBSEB 306A, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
27
|
Abstract
Metformin is currently the first-line drug treatment for type 2 diabetes. Besides its glucose-lowering effect, there is interest in actions of the drug of potential relevance to cardiovascular diseases and cancer. However, the underlying mechanisms of action remain elusive. Convincing data place energy metabolism at the center of metformin's mechanism of action in diabetes and may also be of importance in cardiovascular diseases and cancer. Metformin-induced activation of the energy-sensor AMPK is well documented, but may not account for all actions of the drug. Here, we summarize current knowledge about the different AMPK-dependent and AMPK-independent mechanisms underlying metformin action.
Collapse
Affiliation(s)
- Marc Foretz
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, France
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands; Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Luc Bertrand
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Brussels, Belgium
| | - Michael Pollak
- Department of Oncology, McGill University and Segal Cancer Centre of the Jewish General Hospital, Montreal, Quebec, Canada
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, France.
| |
Collapse
|
28
|
Yao H, Han X, Han X. The cardioprotection of the insulin-mediated PI3K/Akt/mTOR signaling pathway. Am J Cardiovasc Drugs 2014; 14:433-42. [PMID: 25160498 DOI: 10.1007/s40256-014-0089-9] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Apoptosis occurs frequently in myocardial infarction, oxidative stress injury, and ischemia/reperfusion injury, and plays a pivotal role in the development of heart diseases. Inhibition of apoptosis alone does not necessarily lead to meaningful rescue in terms of either cardiomyocyte survival or function. Activation of the PI3K/Akt signaling pathway induced by insulin not only inhibits cardiomyocyte apoptosis but also substantially preserves and even improves regional and overall cardiac function. Insulin can protect cardiomyocytes from apoptosis by regulating a number of signaling molecules, such as eNOS, FOXOs, Bad, GSK-3β, mTOR, NDRG2, and Nrf2, through activating PI3K and Akt. This review focuses on the protective mechanisms and targets of insulin identified in the prevention and treatment of myocardial injury.
Collapse
|
29
|
Balteau M, Van Steenbergen A, Timmermans AD, Dessy C, Behets-Wydemans G, Tajeddine N, Castanares-Zapatero D, Gilon P, Vanoverschelde JL, Horman S, Hue L, Bertrand L, Beauloye C. AMPK activation by glucagon-like peptide-1 prevents NADPH oxidase activation induced by hyperglycemia in adult cardiomyocytes. Am J Physiol Heart Circ Physiol 2014; 307:H1120-33. [DOI: 10.1152/ajpheart.00210.2014] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Exposure of cardiomyocytes to high glucose concentrations (HG) stimulates reactive oxygen species (ROS) production by NADPH oxidase (NOX2). NOX2 activation is triggered by enhanced glucose transport through a sodium-glucose cotransporter (SGLT) but not by a stimulation of glucose metabolism. The aim of this work was to identify potential therapeutic approaches to counteract this glucotoxicity. In cultured adult rat cardiomyocytes incubated with 21 mM glucose (HG), AMP-activated protein kinase (AMPK) activation by A769662 or phenformin nearly suppressed ROS production. Interestingly, glucagon-like peptide 1 (GLP-1), a new antidiabetic drug, concomitantly induced AMPK activation and prevented the HG-mediated ROS production (maximal effect at 100 nM). α2-AMPK, the major isoform expressed in cardiomyocytes (but not α1-AMPK), was activated in response to GLP-1. Anti-ROS properties of AMPK activators were not related to changes in glucose uptake or glycolysis. Using in situ proximity ligation assay, we demonstrated that AMPK activation prevented the HG-induced p47phox translocation to caveolae, whatever the AMPK activators used. NOX2 activation by either α-methyl-d-glucopyranoside, a glucose analog transported through SGLT, or angiotensin II was also counteracted by GLP-1. The crucial role of AMPK in limiting HG-mediated NOX2 activation was demonstrated by overexpressing a constitutively active form of α2-AMPK using adenoviral infection. This overexpression prevented NOX2 activation in response to HG, whereas GLP-1 lost its protective action in α2-AMPK-deficient mouse cardiomyocytes. Under HG, the GLP-1/AMPK pathway inhibited PKC-β2 phosphorylation, a key element mediating p47phox translocation. In conclusion, GLP-1 induces α2-AMPK activation and blocks HG-induced p47phox translocation to the plasma membrane, thereby preventing glucotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Patrick Gilon
- Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique (IREC), and
| | - Jean-Louis Vanoverschelde
- Pôles de 1Recherche Cardiovasculaire,
- Cliniques universitaires Saint Luc, Division of Cardiology, Brussels, Belgium
| | | | - Louis Hue
- de Duve Institute, Université catholique de Louvain (UCL), Brussels
| | | | - Christophe Beauloye
- Pôles de 1Recherche Cardiovasculaire,
- Cliniques universitaires Saint Luc, Division of Cardiology, Brussels, Belgium
| |
Collapse
|
30
|
Maternal undernutrition around the time of conception and embryo number each impact on the abundance of key regulators of cardiac growth and metabolism in the fetal sheep heart. J Dev Orig Health Dis 2014; 4:377-90. [PMID: 24970731 DOI: 10.1017/s2040174413000354] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Poor maternal nutrition before and during pregnancy is associated with an increased risk of cardiovascular disease in later life. To determine the impact of maternal undernutrition during the periconceptional (PCUN: -45 days to 6 days) and preimplantation (PIUN: 0-6 days) periods on cardiac growth and metabolism, we have quantified the mRNA and protein abundance of key regulators of cardiac growth and metabolism in the left ventricle of the sheep fetus in late gestation. The cardiac protein abundance of AMP-activated protein kinase (AMPK), phospho-acetyl CoA carboxykinase (ACC) and pyruvate dehydrogenase kinase-4 (PDK-4) were decreased, whereas ACC was increased in singletons in the PCUN and PIUN groups. In twins, however, cardiac ACC was decreased in the PCUN and PIUN groups, and carnitine palmitoyltransferase-1 (CPT-1) was increased in the PIUN group. In singletons, the cardiac abundance of insulin receptor β (IRβ) was decreased in the PCUN group, and phosphoinositide-dependent protein kinase-1 (PDPK-1) was decreased in the PCUN and PIUN groups. In twins, however, the cardiac abundance of IRβ and phospho-Akt substrate 160kDa (pAS160) were increased in the PIUN group. The cardiac abundance of insulin-like growth factor-2 receptor (IGF-2R), protein kinase C alpha (PKCα) and mammalian target of rapamycin (mTOR) were decreased in PCUN and PIUN singletons and extracellular-signal-regulated kinase (ERK) was also decreased in the PIUN singletons. In contrast, in twins, cardiac abundance of IGF-2R and PKCα were increased in the PCUN and PIUN groups, phospho-ribosomal protein S6 (pRPS6) was increased in the PCUN group, and ERK and eukaryotic initiation factor 4E (eIF4E) were also increased in the PIUN fetuses. In conclusion, maternal undernutrition limited to around the time of conception is sufficient to alter the abundance of key factors regulating cardiac growth and metabolism and this may increase the propensity for cardiovascular diseases in later life.
Collapse
|
31
|
Timmermans AD, Balteau M, Gélinas R, Renguet E, Ginion A, de Meester C, Sakamoto K, Balligand JL, Bontemps F, Vanoverschelde JL, Horman S, Beauloye C, Bertrand L. A-769662 potentiates the effect of other AMP-activated protein kinase activators on cardiac glucose uptake. Am J Physiol Heart Circ Physiol 2014; 306:H1619-30. [PMID: 24748590 DOI: 10.1152/ajpheart.00965.2013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
AMP-activated protein kinase (AMPK), a key cellular sensor of energy, regulates metabolic homeostasis and plays a protective role in the ischemic or diabetic heart. Stimulation of cardiac glucose uptake contributes to this AMPK-mediated protection. The small-molecule AMPK activator A-769662, which binds and directly activates AMPK, has recently been characterized. A-769662-dependent AMPK activation protects the heart against an ischemia-reperfusion episode but is unable to stimulate skeletal muscle glucose uptake. Here, we tried to reconcile these conflicting findings by investigating the impact of A-769662 on cardiac AMPK signaling and glucose uptake. We showed that A-769662 promoted AMPK activation, resulting in the phosphorylation of several downstream targets, but was incapable of stimulating glucose uptake in cultured cardiomyocytes and the perfused heart. The lack of glucose uptake stimulation can be explained by A-769662's narrow specificity, since it selectively activates cardiac AMPK heterotrimeric complexes containing α2/β1-subunits, the others being presumably required for this metabolic outcome. However, when combined with classical AMPK activators, such as metformin, phenformin, oligomycin, or hypoxia, which impact AMPK heterotrimers more broadly via elevation of cellular AMP levels, A-769662 induced more profound AMPK phosphorylation and subsequent glucose uptake stimulation. The synergistic effect of A-769662 under such ischemia-mimetic conditions protected cardiomyocytes against ROS production and cell death. In conclusion, despite the fact that A-769662 activates AMPK, it alone does not significantly stimulate glucose uptake. However, strikingly, its ability of potentiating the action on other AMPK activators makes it a potentially useful participant in the protective role of AMPK in the heart.
Collapse
Affiliation(s)
- Aurélie D Timmermans
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Brussels, Belgium
| | - Magali Balteau
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Brussels, Belgium
| | - Roselle Gélinas
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Brussels, Belgium
| | - Edith Renguet
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Brussels, Belgium
| | - Audrey Ginion
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Brussels, Belgium
| | - Carole de Meester
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Brussels, Belgium
| | - Kei Sakamoto
- Medical Research Council Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Jean-Luc Balligand
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Pharmacology and Therapeutics, Brussels, Belgium; and
| | | | - Jean-Louis Vanoverschelde
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Brussels, Belgium
| | - Sandrine Horman
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Brussels, Belgium
| | - Christophe Beauloye
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Brussels, Belgium
| | - Luc Bertrand
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Brussels, Belgium;
| |
Collapse
|
32
|
Lee CT, Ussher JR, Mohammad A, Lam A, Lopaschuk GD. 5'-AMP-activated protein kinase increases glucose uptake independent of GLUT4 translocation in cardiac myocytes. Can J Physiol Pharmacol 2014; 92:307-14. [PMID: 24708213 DOI: 10.1139/cjpp-2013-0107] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glucose uptake and glycolysis are increased in the heart during ischemia, and this metabolic alteration constitutes an important contributing factor towards ischemic injury. Therefore, it is important to understand glucose uptake regulation in the ischemic heart. There are primarily 2 glucose transporters controlling glucose uptake into cardiac myocytes: GLUT1 and GLUT4. In the non-ischemic heart, insulin stimulates GLUT4 translocation to the sarcolemmal membrane, while both GLUT1 and GLUT4 translocation can occur following AMPK stimulation. Using a newly developed technique involving [(3)H]2-deoxyglucose, we measured glucose uptake in H9c2 ventricular myoblasts, and demonstrated that while insulin has no detectable effect on glucose uptake, phenformin-induced AMPK activation increases glucose uptake 2.5-fold. Furthermore, insulin treatment produced no discernible effect on either Akt serine 473 phosphorylation or AMPKα threonine 172 phosphorylation, while treatment with phenformin results in an increase in AMPKα threonine 172 phosphorylation, and a decrease in Akt serine 473 phosphorylation. Visualization of a dsRed-GLUT4 fusion construct in H9c2 cells by laser confocal microscopy showed that unlike insulin, AMPK activation did not redistribute GLUT4 to the sarcolemmal membrane, suggesting that AMPK may regulate glucose uptake via another glucose transporter. These studies suggest that AMPK is a major regulator of glucose uptake in cardiac myocytes.
Collapse
Affiliation(s)
- Christopher T Lee
- a Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, 8440 112 Street NW, Edmonton, AB T6G 2P4, Canada
| | | | | | | | | |
Collapse
|
33
|
Crosstalk Between Insulin and Toll-like Receptor Signaling Pathways in the Central Nervous system. Mol Neurobiol 2014; 50:797-810. [DOI: 10.1007/s12035-013-8631-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 12/25/2013] [Indexed: 01/04/2023]
|
34
|
Sun A, Cheng Y, Zhang Y, Zhang Q, Wang S, Tian S, Zou Y, Hu K, Ren J, Ge J. Aldehyde dehydrogenase 2 ameliorates doxorubicin-induced myocardial dysfunction through detoxification of 4-HNE and suppression of autophagy. J Mol Cell Cardiol 2014; 71:92-104. [PMID: 24434637 DOI: 10.1016/j.yjmcc.2014.01.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/06/2013] [Accepted: 01/03/2014] [Indexed: 01/24/2023]
Abstract
Mitochondrial aldehyde dehydrogenase (ALDH2) protects against cardiac injury via reducing production of 4-hydroxynonenal (4-HNE) and ROS. This study was designed to examine the impact of ALDH2 on doxorubicin (DOX)-induced cardiomyopathy and mechanisms involved with a focus on autophagy. 4-HNE and autophagic markers were detected by Western blotting in ventricular tissues from normal donors and patients with idiopathic dilated cardiomyopathy. Cardiac function, 4-HNE and levels of autophagic markers were detected in WT, ALDH2 knockout or ALDH2 transfected mice treated with or without DOX. Autophagy regulatory signaling including PI-3K, AMPK and Akt was examined in DOX-treated cardiomyocytes incubated with or without ALDH2 activator Alda-1. DOX-induced myocardial dysfunction, upregulation of 4-HNE and autophagic proteins were further aggravated in ALDH2 knockout mice while they were ameliorated in ALDH2 transfected mice. DOX downregulated Class I and upregulated Class III PI3-kinase, the effect of which was augmented by ALDH2 deletion. Accumulation of 4-HNE and autophagic protein markers in DOX-induced cardiomyocytes was significantly reduced by Alda-1. DOX depressed phosphorylated Akt but not AMPK, the effect was augmented by ALDH2 knockout. The autophagy inhibitor 3-MA attenuated, whereas autophagy inducer rapamycin mimicked DOX-induced cardiomyocyte contractile defects. In addition, rapamycin effectively mitigated Alda-1-offered protective action against DOX-induced cardiomyocyte dysfunction. Our data further revealed downregulated ALDH2 and upregulated autophagy levels in the hearts from patients with dilated cardiomyopathy. Taken together, our findings suggest that inhibition of 4-HNE and autophagy may be a plausible mechanism underscoring ALDH2-offered protection against DOX-induced cardiac defect. This article is part of a Special Issue entitled "Protein Quality Control, the Ubiquitin Proteasome System, and Autophagy".
Collapse
Affiliation(s)
- Aijun Sun
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| | - Yong Cheng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Heart Centre of Zhengzhou Ninth People's Hospital, Zhengzhou, Henan 450000, China
| | - Yingmei Zhang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Qian Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Cardiology, Branch of Shanghai First People's Hospital, Shanghai 200050, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Shan Tian
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Kai Hu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
35
|
Thorn SL, Gollob MH, Harper ME, Beanlands RS, Dekemp RA, Dasilva JN. Chronic AMPK activity dysregulation produces myocardial insulin resistance in the human Arg302Gln-PRKAG2 glycogen storage disease mouse model. EJNMMI Res 2013; 3:48. [PMID: 23829931 PMCID: PMC3707764 DOI: 10.1186/2191-219x-3-48] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 06/27/2013] [Indexed: 11/22/2022] Open
Abstract
Background The cardiac PRKAG2 mutation in the γ2-subunit of adenosine monophosphate activated kinase (AMPK) is characterized by excessive glycogen deposition, hypertrophy, frequent arrhythmias, and progressive conduction system disease. We investigated whether myocardial glucose uptake (MGU) was augmented following insulin stimulation in a mouse model of the PRKAG2 cardiac syndrome. Methods Myocardial and skeletal muscle glucose uptake was assessed with 2-[18F]fluoro-2-deoxyglucose positron emission tomography imaging in n = 3 transgenic wildtype (TGwt) vs n = 7 PRKAG2 mutant (TGmut) mice at baseline and 1 week later, 30 min following acute insulin. Systolic function, cardiac glycogen stores, phospho-AMPK α, and insulin-receptor expression levels were analyzed to corroborate to the in vivo findings. Results TGmut Patlak Ki was reduced 56% at baseline compared to TGwt (0.3 ± 0.2 vs 0.7 ± 0.1, t test p = 0.01). MGU was augmented 71% in TGwt mice following acute insulin from baseline (0.7 ± 0.1 to 1.2 ± 0.2, t test p < 0.05). No change was observed in TGmut mice. As expected for this cardiac specific transgene, skeletal muscle was unaffected at baseline with a 33% to 38% increase (standard uptake values) for both genotypes following insulin stimulation. TGmut mice had a 47% reduction in systolic function with a fourfold increase in cardiac glycogen stores correlated with a 29% reduction in phospho-AMPK α levels. There was no difference in cardiac insulin receptor expression between mouse genotypes. Conclusions These results demonstrate a correlation between insulin resistance and AMPK activity and provide the basis for the use of this animal model for assessing metabolic therapy in the treatment of affected PRKAG2 patients.
Collapse
Affiliation(s)
- Stephanie L Thorn
- National Cardiac PET Centre, Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, Ontario K1Y 4W7, Canada.
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
The main approach to obesity and type-II diabetes is to unravel the mechanisms involved in nutrient absorption and fuel allocation. In conditions of over-nutrition, cells must cope with a multitude of extracellular signals generated by changes in nutrient load, hormonal milieu, adverse cytokine/adipokine profile, and apoptosis/anti-apoptosis processes. To date studies have demonstrate that among all nutrients, lipids and carbohydrates play a major regulatory role in the gene transcription of glycolytic and lipogenic enzymes, insulin, and adipokines. These nutrients mainly exert their effects through the gene expression of sterol responsive binding protein 1 and 2 (SREBP) and the mammalian target of rapamycin (mTOR). Excess of adipose tissue is known to confer a significantly higher risk of coronary artery disease. Administration of rapamycin effectively attenuated inflammation, inhibited progression, and enhanced stability of atherosclerotic plaques in animal models. Herein we discuss the mTOR pathway and the molecular mechanisms of mTOR inhibitors, hypothesizing a possible protective role in atherosclerosis, taking into account also previous clinical studies emphasizing their opposite role.
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Italy.
| | | |
Collapse
|
37
|
Demeulder B, Zarrinpashneh E, Ginion A, Viollet B, Hue L, Rider MH, Vanoverschelde JL, Beauloye C, Horman S, Bertrand L. Differential regulation of eEF2 and p70S6K by AMPKalpha2 in heart. Biochim Biophys Acta Mol Basis Dis 2013; 1832:780-90. [DOI: 10.1016/j.bbadis.2013.02.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 02/17/2013] [Accepted: 02/25/2013] [Indexed: 01/13/2023]
|
38
|
Nagendran J, Waller TJ, Dyck JRB. AMPK signalling and the control of substrate use in the heart. Mol Cell Endocrinol 2013; 366:180-93. [PMID: 22750050 DOI: 10.1016/j.mce.2012.06.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 03/29/2012] [Accepted: 06/21/2012] [Indexed: 12/21/2022]
Abstract
All mammalian cells rely on adenosine triphosphate (ATP) to maintain function and for survival. The heart has the highest basal ATP demand of any organ due to the necessity for continuous contraction. As such, the ability of the cardiomyocyte to monitor cellular energy status and adapt the supply of substrates to match the energy demand is crucial. One important serine/threonine protein kinase that monitors cellular energy status in the heart is adenosine monophosphate activated protein kinase (AMPK). AMPK is also a key enzyme that controls multiple catabolic and anabolic biochemical pathways in the heart and indirectly plays a crucial role in regulating cardiac function in both physiological and pathophysiological conditions. Herein, we review the involvement of AMPK in myocardial fatty acid and glucose transport and utilization, as it relates to basal cardiac function. We also assess the literature amassed on cardiac AMPK and discuss the controversies surrounding the role of AMPK in physiological and pathophysiological processes in the heart. The work reviewed herein also emphasizes areas that require further investigation for the purpose of eventually translating this information into improved patient care.
Collapse
Affiliation(s)
- Jeevan Nagendran
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
39
|
CD36 inhibition prevents lipid accumulation and contractile dysfunction in rat cardiomyocytes. Biochem J 2013; 448:43-53. [PMID: 22780108 DOI: 10.1042/bj20120060] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
An increased cardiac fatty acid supply and increased sarcolemmal presence of the long-chain fatty acid transporter CD36 are associated with and contribute to impaired cardiac insulin sensitivity and function. In the present study we aimed at preventing the development of insulin resistance and contractile dysfunction in cardiomyocytes by blocking CD36-mediated palmitate uptake. Insulin resistance and contractile dysfunction were induced in primary cardiomyocytes by 48 h incubation in media containing either 100 nM insulin (high insulin; HI) or 200 μM palmitate (high palmitate; HP). Under both culture conditions, insulin-stimulated glucose uptake and Akt phosphorylation were abrogated or markedly reduced. Furthermore, cardiomyocytes cultured in each medium displayed elevated sarcolemmal CD36 content, increased basal palmitate uptake, lipid accumulation and decreased sarcomere shortening. Immunochemical CD36 inhibition enhanced basal glucose uptake and prevented elevated basal palmitate uptake, triacylglycerol accumulation and contractile dysfunction in cardiomyocytes cultured in either medium. Additionally, CD36 inhibition prevented loss of insulin signalling in cells cultured in HP, but not in HI medium. In conclusion, CD36 inhibition prevents lipid accumulation and lipid-induced contractile dysfunction in cardiomyocytes, but probably independently of effects on insulin signalling. Nonetheless, pharmacological CD36 inhibition may be considered as a treatment strategy to counteract impaired functioning of the lipid-loaded heart.
Collapse
|
40
|
Steinbusch LKM, Dirkx E, Hoebers NTH, Roelants V, Foretz M, Viollet B, Diamant M, van Eys G, Ouwens DM, Bertrand L, Glatz JFC, Luiken JJFP. Overexpression of AMP-activated protein kinase or protein kinase D prevents lipid-induced insulin resistance in cardiomyocytes. J Mol Cell Cardiol 2012; 55:165-73. [PMID: 23159540 DOI: 10.1016/j.yjmcc.2012.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 11/01/2012] [Accepted: 11/06/2012] [Indexed: 01/13/2023]
Abstract
During lipid oversupply, the heart becomes insulin resistant, as exemplified by defective insulin-stimulated glucose uptake, and will develop diastolic dysfunction. In the healthy heart, not only insulin, but also increased contractile activity stimulates glucose uptake. Upon increased contraction both AMP-activated protein kinase (AMPK) and protein kinase D (PKD) are activated, and mediate the stimulation of glucose uptake into cardiomyocytes. Therefore, each of these kinases is a potential therapeutic target in the diabetic heart because they may serve to bypass defective insulin-stimulated glucose uptake. To test the preventive potential of these kinases against loss of insulin-stimulated glucose uptake, AMPK or PKD were adenovirally overexpressed in primary cultures of insulin resistant cardiomyocytes for assaying substrate uptake, insulin responsiveness and lipid accumulation. To induce insulin resistance and lipid loading, rat primary cardiomyocytes were cultured in the presence of high insulin (100 nM; HI) or high palmitate (palmitate/BSA: 3/1; HP). HI and HP each reduced insulin responsiveness, and increased basal palmitate uptake and lipid storage. Overexpression of each of the kinases prevented loss of insulin-stimulated glucose uptake. Overexpression of AMPK also prevented loss of insulin signaling in HI- and HP-cultured cardiomyocytes, but did not prevent lipid accumulation. In contrast, overexpression of PKD prevented lipid accumulation, but not loss of insulin signaling in HI- and HP-cultured cardiomyocytes. In conclusion, AMPK and PKD prevent loss of insulin-stimulated glucose uptake into cardiomyocytes cultured under insulin resistance-inducing conditions through different mechanisms. This article is part of a Special Issue entitled "Focus on Cardiac Metabolism".
Collapse
|
41
|
Zismanov V, Drucker L, Attar-Schneider O, Matalon ST, Pasmanik-Chor M, Lishner M. Tetraspanins stimulate protein synthesis in myeloma cell lines. J Cell Biochem 2012; 113:2500-10. [PMID: 22415769 DOI: 10.1002/jcb.24126] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Intensive protein synthesis is a unique and differential trait of multiple myeloma (MM) cells. Previously we showed that tetraspanin (CD81, CD82) overexpression in MM cell lines attenuated Akt/mTOR cascades, activated UPR, and caused autophagic death, suggesting breach of protein homeostasis. Here, we explored the role of protein synthesis in the tetraspanin-induced MM cell death. Contrary to attenuation of the major metabolic regulator, mTOR we determined elevated steady-state levels of protein in CD81N1/CD82N1 transfected MM lines (RPMI-8226, CAG). Elevated levels of immunoglobulins supported increased protein production in RPMI-8226. Changes in cell morphology consistent with elevated protein synthesis were also determined (cell, nuclei, and nucleoli sizes and ratios). Increased levels of phospho-rpS6 and decreased levels of phospho-AMPK were consistent with increased translation but independent of mTOR. Involvement of p38 and its role in tetraspanin induced translation and cell death were demonstrated. Microarray analyses of tetraspanin transfected MM cell lines revealed activation of protein synthesis signaling cascades and signals implicated in ribosome biogenesis (snoRNAs). Finally, we showed tetraspanins elevated protein synthesis was instrumental to MM cells' death. This work explores and demonstrates that excessive protein translation can be detrimental to MM cell lines and therefore may present a therapeutic target. Proteostasis is particularly important in MM because it integrates the high levels of protein production unique to myeloma cells with critically important microenvironmental cues. We suggest that increasing translation may be the path of least resistance in MM and thus may afford a novel platform for strategically designed therapy.
Collapse
|
42
|
Montessuit C, Lerch R. Regulation and dysregulation of glucose transport in cardiomyocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:848-56. [PMID: 22967513 DOI: 10.1016/j.bbamcr.2012.08.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 08/08/2012] [Accepted: 08/10/2012] [Indexed: 11/29/2022]
Abstract
The ability of the heart muscle to derive energy from a wide variety of substrates provides the myocardium with remarkable capacity to adapt to the ever-changing metabolic environment depending on factors including nutritional state and physical activity. There is increasing evidence that loss of metabolic flexibility of the myocardium contributes to cardiac dysfunction in disease conditions such as diabetes, ischemic heart disease and heart failure. At the level of glucose metabolism reduced metabolic adaptation in most cases is characterized by impaired stimulation of transarcolemmal glucose transport in the cardiomyocytes in response to insulin, referred to as insulin resistance, or to other stimuli such as energy deficiency. This review discusses cellular mechanisms involved in the regulation of glucose uptake in cardiomyocytes and their potential implication in impairment of stimulation of glucose transport under disease conditions. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.
Collapse
Affiliation(s)
- Christophe Montessuit
- Department of Medical Specialties, Geneva University Hospitals, Geneva, Switzerland.
| | | |
Collapse
|
43
|
Horman S, Beauloye C, Vanoverschelde JL, Bertrand L. AMP-activated Protein Kinase in the Control of Cardiac Metabolism and Remodeling. Curr Heart Fail Rep 2012; 9:164-73. [DOI: 10.1007/s11897-012-0102-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
44
|
Asrih M, Lerch R, Papageorgiou I, Pellieux C, Montessuit C. Differential regulation of stimulated glucose transport by free fatty acids and PPARα or -δ agonists in cardiac myocytes. Am J Physiol Endocrinol Metab 2012; 302:E872-84. [PMID: 22297301 DOI: 10.1152/ajpendo.00427.2011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Stimulation of glucose transport in response to insulin or metabolic stress is an important determinant of cardiac myocyte function and survival, particularly during ischemia-reperfusion episodes. The impact of dyslipidemia and its consequence PPAR activation on stimulated glucose transport in cardiac myocytes remains unknown. Isolated adult rat cardiac myocytes were chronically exposed to free fatty acids (FFA) or PPAR agonists. Insulin- (ISGT) and oligomycin-stimulated glucose transport (OSGT) and related cell signaling were analyzed. Exposure of cardiac myocytes to FFA reduced both ISGT and OSGT. Exposure to either PPARα or PPARδ agonists, but not to a PPARγ agonist, reduced ISGT but not OSGT and increased fatty acid oxidation (FAO). The reduction in ISGT was associated with impaired insulin signaling and, in the case of PPAR stimulation, overexpression of SOCS-3, a protein known to hinder proximal insulin signaling. In contrast, the reduction of OSGT could not be explained by a reduced activity of the cellular energy-sensing system, as assessed from the maintained phosphorylation state of AMPK. Inhibition of FAO at the level of mitochondrial acylcarnitine uptake restored OSGT but not ISGT. Seemingly paradoxically, further stimulation of FAO with PPARα or PPARδ agonists also restored OSGT but not ISGT. Together, these results suggest that inhibition of OSGT occurs downstream of energy gauging and is caused by some intermediate(s) of fatty acid oxidation, which does not appear to be acylcarnitines. The results indicate that the mechanisms underlying FFA-mediated inhibition of ISGT and OSGT differ remarkably.
Collapse
MESH Headings
- Animals
- Antimetabolites/metabolism
- Biological Transport, Active/drug effects
- Blotting, Western
- Cells, Cultured
- Deoxyglucose/metabolism
- Fatty Acids, Nonesterified/pharmacology
- Glucose/metabolism
- Glucose Transport Proteins, Facilitative/biosynthesis
- Glucose Transport Proteins, Facilitative/genetics
- Hypoglycemic Agents/pharmacology
- Insulin/pharmacology
- Male
- Microscopy, Confocal
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Oligomycins/pharmacology
- Oxidation-Reduction
- PPAR alpha/agonists
- PPAR delta/agonists
- Palmitates/metabolism
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Real-Time Polymerase Chain Reaction
- Signal Transduction/drug effects
- Uncoupling Agents/pharmacology
Collapse
Affiliation(s)
- Mohamed Asrih
- Division of Cardiology, Department of Medical Specialties, Geneva University Hospitals, Switzerland
| | | | | | | | | |
Collapse
|
45
|
Ma L, Gul R, Habibi J, Yang M, Pulakat L, Whaley-Connell A, Ferrario CM, Sowers JR. Nebivolol improves diastolic dysfunction and myocardial remodeling through reductions in oxidative stress in the transgenic (mRen2) rat. Am J Physiol Heart Circ Physiol 2012; 302:H2341-51. [PMID: 22447938 DOI: 10.1152/ajpheart.01126.2011] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Angiotensin II contributes to myocardial tissue remodeling and interstitial fibrosis through NADPH oxidase-mediated generation of oxidative stress in the progression of heart failure. Recent data have suggested that nebivolol, a third-generation β-blocker, improves diastolic dysfunction by targeting nitric oxide (NO) and metabolic pathways that decrease interstitial fibrosis. We sought to determine if targeting NO would improve diastolic function in a model of tissue renin-angiotensin system overactivation. We used the transgenic (TG) (mRen2)27 rat, which overexpresses the murine renin transgene and manifests insulin resistance and left ventricular dysfunction. We treated 6- to 7-wk-old TG (mRen2)27 rats and age-matched Sprague-Dawley control rats with nebivolol (10 mg·kg(-1)·day(-1)) or placebo via osmotic minipumps for a period of 21 days. Compared with Sprague-Dawley control rats, TG (mRen2)27 rats displayed a prolonged diastolic relaxation time and reduced initial filling rate associated with increased interstitial fibrosis and left ventricular hypertrophy. These findings were temporally related to increased NADPH oxidase activity and subunits p47(phox) and Rac1 and increased total ROS and peroxynitrite formation in parallel with reductions in the antioxidant heme oxygenase as well as the phosphorylation/activation of endothelial NO synthase and PKB/Akt. Treatment with nebivolol restored diastolic function and interstitial fibrosis through increases in the phosphorylation of 5'-AMP-activated protein kinase, Akt, and endothelial NO synthase and reductions in oxidant stress. These results support that targeting NO with nebivolol treatment improves diastolic dysfunction through reducing myocardial oxidative stress by enhancing 5'-AMP-activated protein kinase and Akt activation of NO biosynthesis.
Collapse
Affiliation(s)
- Lixin Ma
- Department of Radiology, University of Missouri School of Medicine, Columbia, USA
| | | | | | | | | | | | | | | |
Collapse
|