1
|
Dodd MS, Ambrose L, Ball V, Clarke K, Carr CA, Tyler DJ. The age-dependent development of abnormal cardiac metabolism in the peroxisome proliferator-activated receptor α-knockout mouse. Atherosclerosis 2024; 399:118599. [PMID: 39307613 DOI: 10.1016/j.atherosclerosis.2024.118599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND AND AIMS Peroxisome proliferator-activated receptor α (PPARα) is crucial for regulating cardiac β-oxidation in the heart, liver, and kidney. Ageing can induce cardiac metabolic alterations, but the role of PPARα has not been extensively characterised. The aim of this research was to investigate the role of PPARα in the aged heart. METHODS Hyperpolarized [1-13C]pyruvate was used to evaluate in vivo cardiac carbohydrate metabolism in fed and fasted young (3 months) and old (20-22 months) PPARα knockout (KO) mice versus controls. Cine MRI assessed cardiac structural and functional changes. Cardiac tissue analysis included qRT-PCR and Western blotting for Pparα, medium chain acyl-CoA dehydrenase (MCAD), uncoupling protein (UCP) 3, glucose transporter (GLUT) 4 and PDH kinase (PDK) 1,2, and 4 expression. RESULTS PPARα-KO hearts from both young and old mice showed significantly reduced Pparα mRNA and a 58-59 % decrease in MCAD protein levels compared to controls. Cardiac PDH flux was similar in young control and PPARα-KO mice but 96 % higher in old PPARα-KO mice. Differences between genotypes were consistent in fed and fasted states, with reduced PDH flux when fasted. Increased PDH flux was accompanied by a 179 % rise in myocardial GLUT4 protein. No differences in PDK 1, 2, or 4 protein levels were observed between fed groups, indicating the increased PDH flux in aged PPARα-KO mice was not due to changes in PDH phosphorylation. CONCLUSIONS Aged PPARα-KO mice demonstrated higher cardiac PDH flux compared to controls, facilitated by increased myocardial GLUT4 protein levels, leading to enhanced glucose uptake and glycolysis.
Collapse
Affiliation(s)
- Michael S Dodd
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom; Centre for Health and Life Sciences, Coventry University, Coventry, United Kingdom.
| | - Lucy Ambrose
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| | - Vicky Ball
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| | - Kieran Clarke
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| | - Carolyn A Carr
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| | - Damian J Tyler
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| |
Collapse
|
2
|
Kociper B, Škorja Milić N, Ogrizek I, Miš K, Pirkmajer S. Inhibition of the ubiquitin-proteasome system reduces the abundance of pyruvate dehydrogenase kinase 1 in cultured myotubes. J Muscle Res Cell Motil 2024; 45:155-169. [PMID: 39080182 DOI: 10.1007/s10974-024-09679-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/05/2024] [Indexed: 08/11/2024]
Abstract
Pyruvate dehydrogenase kinase (PDK), which phosphorylates the pyruvate dehydrogenase complex, regulates glucose metabolism in skeletal muscle. PDK1, an isozyme whose expression is controlled by hypoxia-inducible factor-1α (HIF-1α), is thought to play a role in muscle adaptation to hypoxia. While transcriptional upregulation of PDK1 by HIF-1α is well characterised, mechanisms controlling proteolysis of PDK1 in skeletal muscle have not been thoroughly investigated. Proteasome inhibitor MG132 paradoxically reduced the abundance of PDK1 in human cancer cells and rat L6 myotubes, suggesting that MG132 might direct PDK1 towards autophagic degradation. The objectives of our current study were to determine (1) whether MG132 suppresses PDK1 levels in primary human myotubes, (2) whether chloroquine, an inhibitor of autophagy, prevents MG132-induced suppression of PDK1 in L6 myotubes, and (3) whether PYR-41, an inhibitor of ubiquitination, suppresses PDK1 in L6 myotubes. Using qPCR and/or immunoblotting, we found that despite markedly upregulating HIF-1α protein, MG132 did not alter the PDK1 expression in cultured primary human myotubes, while it suppressed both PDK1 mRNA and protein in L6 myotubes. The PDK1 levels in L6 myotubes were suppressed also during co-treatment with chloroquine and MG132. PYR-41 markedly increased the abundance of HIF-1α in primary human and L6 myotubes, while reducing the abundance of PDK1. In L6 myotubes treated with PYR-41, chloroquine increased the abundance of the epidermal growth factor receptor, but did not prevent the suppression of PDK1. Collectively, our results suggest that cultured myotubes degrade PDK1 via a pathway that cannot be inhibited by MG132, PYR-41, and/or chloroquine.
Collapse
Affiliation(s)
- Blaž Kociper
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| | - Nives Škorja Milić
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| | - Ivana Ogrizek
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| | - Katarina Miš
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| | - Sergej Pirkmajer
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia.
| |
Collapse
|
3
|
Rajab SAS, Andersen LK, Kenter LW, Berlinsky DL, Borski RJ, McGinty AS, Ashwell CM, Ferket PR, Daniels HV, Reading BJ. Combinatorial metabolomic and transcriptomic analysis of muscle growth in hybrid striped bass (female white bass Morone chrysops x male striped bass M. saxatilis). BMC Genomics 2024; 25:580. [PMID: 38858615 PMCID: PMC11165755 DOI: 10.1186/s12864-024-10325-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 04/19/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Understanding growth regulatory pathways is important in aquaculture, fisheries, and vertebrate physiology generally. Machine learning pattern recognition and sensitivity analysis were employed to examine metabolomic small molecule profiles and transcriptomic gene expression data generated from liver and white skeletal muscle of hybrid striped bass (white bass Morone chrysops x striped bass M. saxatilis) representative of the top and bottom 10 % by body size of a production cohort. RESULTS Larger fish (good-growth) had significantly greater weight, total length, hepatosomatic index, and specific growth rate compared to smaller fish (poor-growth) and also had significantly more muscle fibers of smaller diameter (≤ 20 µm diameter), indicating active hyperplasia. Differences in metabolomic pathways included enhanced energetics (glycolysis, citric acid cycle) and amino acid metabolism in good-growth fish, and enhanced stress, muscle inflammation (cortisol, eicosanoids) and dysfunctional liver cholesterol metabolism in poor-growth fish. The majority of gene transcripts identified as differentially expressed between groups were down-regulated in good-growth fish. Several molecules associated with important growth-regulatory pathways were up-regulated in muscle of fish that grew poorly: growth factors including agt and agtr2 (angiotensins), nicotinic acid (which stimulates growth hormone production), gadd45b, rgl1, zfp36, cebpb, and hmgb1; insulin-like growth factor signaling (igfbp1 and igf1); cytokine signaling (socs3, cxcr4); cell signaling (rgs13, rundc3a), and differentiation (rhou, mmp17, cd22, msi1); mitochondrial uncoupling proteins (ucp3, ucp2); and regulators of lipid metabolism (apoa1, ldlr). Growth factors pttg1, egfr, myc, notch1, and sirt1 were notably up-regulated in muscle of good-growing fish. CONCLUSION A combinatorial pathway analysis using metabolomic and transcriptomic data collectively suggested promotion of cell signaling, proliferation, and differentiation in muscle of good-growth fish, whereas muscle inflammation and apoptosis was observed in poor-growth fish, along with elevated cortisol (an anti-inflammatory hormone), perhaps related to muscle wasting, hypertrophy, and inferior growth. These findings provide important biomarkers and mechanisms by which growth is regulated in fishes and other vertebrates as well.
Collapse
Affiliation(s)
- Sarah A S Rajab
- Department of Applied Ecology, North Carolina State University, 100 Eugene Brooks Avenue, Box 7617, Raleigh, NC, 27695, USA
| | - Linnea K Andersen
- Department of Applied Ecology, North Carolina State University, 100 Eugene Brooks Avenue, Box 7617, Raleigh, NC, 27695, USA
| | - Linas W Kenter
- Department of Agriculture, Nutrition, and Food Systems, University of New Hampshire, Durham, NH, USA
| | - David L Berlinsky
- Department of Agriculture, Nutrition, and Food Systems, University of New Hampshire, Durham, NH, USA
| | - Russell J Borski
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Andrew S McGinty
- North Carolina State University, Pamlico Aquaculture Field Laboratory, Aurora, NC, USA
| | - Christopher M Ashwell
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, USA
| | - Peter R Ferket
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, USA
| | - Harry V Daniels
- Department of Applied Ecology, North Carolina State University, 100 Eugene Brooks Avenue, Box 7617, Raleigh, NC, 27695, USA
| | - Benjamin J Reading
- Department of Applied Ecology, North Carolina State University, 100 Eugene Brooks Avenue, Box 7617, Raleigh, NC, 27695, USA.
- North Carolina State University, Pamlico Aquaculture Field Laboratory, Aurora, NC, USA.
| |
Collapse
|
4
|
Maurissen TL, Spielmann AJ, Schellenberg G, Bickle M, Vieira JR, Lai SY, Pavlou G, Fauser S, Westenskow PD, Kamm RD, Ragelle H. Modeling early pathophysiological phenotypes of diabetic retinopathy in a human inner blood-retinal barrier-on-a-chip. Nat Commun 2024; 15:1372. [PMID: 38355716 PMCID: PMC10866954 DOI: 10.1038/s41467-024-45456-z] [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: 03/17/2023] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
Diabetic retinopathy (DR) is a microvascular disorder characterized by inner blood-retinal barrier (iBRB) breakdown and irreversible vision loss. While the symptoms of DR are known, disease mechanisms including basement membrane thickening, pericyte dropout and capillary damage remain poorly understood and interventions to repair diseased iBRB microvascular networks have not been developed. In addition, current approaches using animal models and in vitro systems lack translatability and predictivity to finding new target pathways. Here, we develop a diabetic iBRB-on-a-chip that produces pathophysiological phenotypes and disease pathways in vitro that are representative of clinical diagnoses. We show that diabetic stimulation of the iBRB-on-a-chip mirrors DR features, including pericyte loss, vascular regression, ghost vessels, and production of pro-inflammatory factors. We also report transcriptomic data from diabetic iBRB microvascular networks that may reveal drug targets, and examine pericyte-endothelial cell stabilizing strategies. In summary, our model recapitulates key features of disease, and may inform future therapies for DR.
Collapse
Affiliation(s)
- Thomas L Maurissen
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Alena J Spielmann
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Gabriella Schellenberg
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Marc Bickle
- Roche Pharma Research and Early Development, Institute of Human Biology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jose Ricardo Vieira
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Si Ying Lai
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Georgios Pavlou
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sascha Fauser
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Peter D Westenskow
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Héloïse Ragelle
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| |
Collapse
|
5
|
Gopal K, Abdualkader AM, Li X, Greenwell AA, Karwi QG, Altamimi TR, Saed C, Uddin GM, Darwesh AM, Jamieson KL, Kim R, Eaton F, Seubert JM, Lopaschuk GD, Ussher JR, Al Batran R. Loss of muscle PDH induces lactic acidosis and adaptive anaplerotic compensation via pyruvate-alanine cycling and glutaminolysis. J Biol Chem 2023; 299:105375. [PMID: 37865313 PMCID: PMC10692893 DOI: 10.1016/j.jbc.2023.105375] [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: 07/07/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 10/23/2023] Open
Abstract
Pyruvate dehydrogenase (PDH) is the rate-limiting enzyme for glucose oxidation that links glycolysis-derived pyruvate with the tricarboxylic acid (TCA) cycle. Although skeletal muscle is a significant site for glucose oxidation and is closely linked with metabolic flexibility, the importance of muscle PDH during rest and exercise has yet to be fully elucidated. Here, we demonstrate that mice with muscle-specific deletion of PDH exhibit rapid weight loss and suffer from severe lactic acidosis, ultimately leading to early mortality under low-fat diet provision. Furthermore, loss of muscle PDH induces adaptive anaplerotic compensation by increasing pyruvate-alanine cycling and glutaminolysis. Interestingly, high-fat diet supplementation effectively abolishes early mortality and rescues the overt metabolic phenotype induced by muscle PDH deficiency. Despite increased reliance on fatty acid oxidation during high-fat diet provision, loss of muscle PDH worsens exercise performance and induces lactic acidosis. These observations illustrate the importance of muscle PDH in maintaining metabolic flexibility and preventing the development of metabolic disorders.
Collapse
Affiliation(s)
- Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Abdualrahman Mohammed Abdualkader
- Faculty of Pharmacy, Université de Montréal, Montréal, Quebec, Canada; Montreal Diabetes Research Center, Montréal, Quebec, Canada; Cardiometabolic Health, Diabetes and Obesity Research Network, Montréal, Quebec, Canada
| | - Xiaobei Li
- Faculty of Pharmacy, Université de Montréal, Montréal, Quebec, Canada; Montreal Diabetes Research Center, Montréal, Quebec, Canada; Cardiometabolic Health, Diabetes and Obesity Research Network, Montréal, Quebec, Canada
| | - Amanda A Greenwell
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Qutuba G Karwi
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada; Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, Newfoundland and Labrador, Canada
| | - Tariq R Altamimi
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Christina Saed
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Golam M Uddin
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Ahmed M Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - K Lockhart Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Ryekjang Kim
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Farah Eaton
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Rami Al Batran
- Faculty of Pharmacy, Université de Montréal, Montréal, Quebec, Canada; Montreal Diabetes Research Center, Montréal, Quebec, Canada; Cardiometabolic Health, Diabetes and Obesity Research Network, Montréal, Quebec, Canada.
| |
Collapse
|
6
|
Zhao Y, Xiong W, Li C, Zhao R, Lu H, Song S, Zhou Y, Hu Y, Shi B, Ge J. Hypoxia-induced signaling in the cardiovascular system: pathogenesis and therapeutic targets. Signal Transduct Target Ther 2023; 8:431. [PMID: 37981648 PMCID: PMC10658171 DOI: 10.1038/s41392-023-01652-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 11/21/2023] Open
Abstract
Hypoxia, characterized by reduced oxygen concentration, is a significant stressor that affects the survival of aerobic species and plays a prominent role in cardiovascular diseases. From the research history and milestone events related to hypoxia in cardiovascular development and diseases, The "hypoxia-inducible factors (HIFs) switch" can be observed from both temporal and spatial perspectives, encompassing the occurrence and progression of hypoxia (gradual decline in oxygen concentration), the acute and chronic manifestations of hypoxia, and the geographical characteristics of hypoxia (natural selection at high altitudes). Furthermore, hypoxia signaling pathways are associated with natural rhythms, such as diurnal and hibernation processes. In addition to innate factors and natural selection, it has been found that epigenetics, as a postnatal factor, profoundly influences the hypoxic response and progression within the cardiovascular system. Within this intricate process, interactions between different tissues and organs within the cardiovascular system and other systems in the context of hypoxia signaling pathways have been established. Thus, it is the time to summarize and to construct a multi-level regulatory framework of hypoxia signaling and mechanisms in cardiovascular diseases for developing more therapeutic targets and make reasonable advancements in clinical research, including FDA-approved drugs and ongoing clinical trials, to guide future clinical practice in the field of hypoxia signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Weidong Xiong
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China
| | - Chaofu Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - Junbo Ge
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
7
|
Teaney NA, Cyr NE. FoxO1 as a tissue-specific therapeutic target for type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1286838. [PMID: 37941908 PMCID: PMC10629996 DOI: 10.3389/fendo.2023.1286838] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023] Open
Abstract
Forkhead box O (FoxO) proteins are transcription factors that mediate many aspects of physiology and thus have been targeted as therapeutics for several diseases including metabolic disorders such as type 2 diabetes mellitus (T2D). The role of FoxO1 in metabolism has been well studied, but recently FoxO1's potential for diabetes prevention and therapy has been debated. For example, studies have shown that increased FoxO1 activity in certain tissue types contributes to T2D pathology, symptoms, and comorbidities, yet in other tissue types elevated FoxO1 has been reported to alleviate symptoms associated with diabetes. Furthermore, studies have reported opposite effects of active FoxO1 in the same tissue type. For example, in the liver, FoxO1 contributes to T2D by increasing hepatic glucose production. However, FoxO1 has been shown to either increase or decrease hepatic lipogenesis as well as adipogenesis in white adipose tissue. In skeletal muscle, FoxO1 reduces glucose uptake and oxidation, promotes lipid uptake and oxidation, and increases muscle atrophy. While many studies show that FoxO1 lowers pancreatic insulin production and secretion, others show the opposite, especially in response to oxidative stress and inflammation. Elevated FoxO1 in the hypothalamus increases the risk of developing T2D. However, increased FoxO1 may mitigate Alzheimer's disease, a neurodegenerative disease strongly associated with T2D. Conversely, accumulating evidence implicates increased FoxO1 with Parkinson's disease pathogenesis. Here we review FoxO1's actions in T2D conditions in metabolic tissues that abundantly express FoxO1 and highlight some of the current studies targeting FoxO1 for T2D treatment.
Collapse
Affiliation(s)
- Nicole A. Teaney
- Stonehill College, Neuroscience Program, Easton, MA, United States
| | - Nicole E. Cyr
- Stonehill College, Neuroscience Program, Easton, MA, United States
- Stonehill College, Department of Biology, Easton, MA, United States
| |
Collapse
|
8
|
Wu G, Baumeister R, Heimbucher T. Molecular Mechanisms of Lipid-Based Metabolic Adaptation Strategies in Response to Cold. Cells 2023; 12:1353. [PMID: 37408188 PMCID: PMC10216534 DOI: 10.3390/cells12101353] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/24/2023] [Accepted: 05/05/2023] [Indexed: 07/07/2023] Open
Abstract
Temperature changes and periods of detrimental cold occur frequently for many organisms in their natural habitats. Homeothermic animals have evolved metabolic adaptation strategies to increase mitochondrial-based energy expenditure and heat production, largely relying on fat as a fuel source. Alternatively, certain species are able to repress their metabolism during cold periods and enter a state of decreased physiological activity known as torpor. By contrast, poikilotherms, which are unable to maintain their internal temperature, predominantly increase membrane fluidity to diminish cold-related damage from low-temperature stress. However, alterations of molecular pathways and the regulation of lipid-metabolic reprogramming during cold exposure are poorly understood. Here, we review organismal responses that adjust fat metabolism during detrimental cold stress. Cold-related changes in membranes are detected by membrane-bound sensors, which signal to downstream transcriptional effectors, including nuclear hormone receptors of the PPAR (peroxisome proliferator-activated receptor) subfamily. PPARs control lipid metabolic processes, such as fatty acid desaturation, lipid catabolism and mitochondrial-based thermogenesis. Elucidating the underlying molecular mechanisms of cold adaptation may improve beneficial therapeutic cold treatments and could have important implications for medical applications of hypothermia in humans. This includes treatment strategies for hemorrhagic shock, stroke, obesity and cancer.
Collapse
Affiliation(s)
- Gang Wu
- Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Ralf Baumeister
- Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Center for Biochemistry and Molecular Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Thomas Heimbucher
- Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
9
|
Kim MJ, Sinam IS, Siddique Z, Jeon JH, Lee IK. The Link between Mitochondrial Dysfunction and Sarcopenia: An Update Focusing on the Role of Pyruvate Dehydrogenase Kinase 4. Diabetes Metab J 2023; 47:153-163. [PMID: 36635027 PMCID: PMC10040620 DOI: 10.4093/dmj.2022.0305] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/13/2022] [Indexed: 01/14/2023] Open
Abstract
Sarcopenia, defined as a progressive loss of muscle mass and function, is typified by mitochondrial dysfunction and loss of mitochondrial resilience. Sarcopenia is associated not only with aging, but also with various metabolic diseases characterized by mitochondrial dyshomeostasis. Pyruvate dehydrogenase kinases (PDKs) are mitochondrial enzymes that inhibit the pyruvate dehydrogenase complex, which controls pyruvate entry into the tricarboxylic acid cycle and the subsequent adenosine triphosphate production required for normal cellular activities. PDK4 is upregulated in mitochondrial dysfunction-related metabolic diseases, especially pathologic muscle conditions associated with enhanced muscle proteolysis and aberrant myogenesis. Increases in PDK4 are associated with perturbation of mitochondria-associated membranes and mitochondrial quality control, which are emerging as a central mechanism in the pathogenesis of metabolic disease-associated muscle atrophy. Here, we review how mitochondrial dysfunction affects sarcopenia, focusing on the role of PDK4 in mitochondrial homeostasis. We discuss the molecular mechanisms underlying the effects of PDK4 on mitochondrial dysfunction in sarcopenia and show that targeting mitochondria could be a therapeutic target for treating sarcopenia.
Collapse
Affiliation(s)
- Min-Ji Kim
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Ibotombi Singh Sinam
- Bio-Medical Research Institute, Kyungpook National University Hospital, Daegu, Korea
| | - Zerwa Siddique
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Korea
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Korea
| | - Jae-Han Jeon
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
- Corresponding author: In-Kyu Lee https://orcid.org/0000-0002-2261-7269 Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu 41944, Korea E-mail:
| |
Collapse
|
10
|
Peng G, Zhu C, Sun Q, Li J, Chen Y, Guo Y, Ji H, Yang F, Dong W. Testicular miRNAs and tsRNAs provide insight into gene regulation during overwintering and reproduction of Onychostoma macrolepis. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:481-499. [PMID: 35595880 DOI: 10.1007/s10695-022-01078-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/19/2022] [Indexed: 06/15/2023]
Abstract
The late overwintering period and breeding period are two important developmental stages of testis in Onychostoma macrolepis. Small non-coding RNAs (sncRNAs) are well-known regulators of biological processes associated with numerous biological processes. This study aimed to elucidate the roles of four sncRNA classes (microRNAs [miRNAs], Piwi-interacting RNAs [piRNAs], tRNA-derived small RNAs [tsRNAs], and rRNA-derived small RNAs [rsRNAs]) across testes in the late overwintering period (in March) and breeding period (in June) by high-throughput sequencing. The testis of O. macrolepis displayed the highest levels of piRNAs and lowest levels of rsRNAs. Compared with miRNAs and tsRNAs in June, tsRNAs in March had a higher abundance, while miRNAs in March had a much lower abundance. Bioinformatics analysis identified 1,362 and 1,340 differentially expressed miRNAs and tsRNAs, respectively. Further analysis showed that miR-200-1, miR-143-1, tRFi-Lys-CTT-1, and tRFi-Glu-CTC-1 could play critical roles during the overwintering and breeding periods. Our findings provided an unprecedented insight to reveal the epigenetic mechanism underlying the overwintering and reproduction process of male O. macrolepis.
Collapse
Affiliation(s)
- Guofan Peng
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
- Biology Research Centre of Qin Mountains Wildlife, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Chao Zhu
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
- Biology Research Centre of Qin Mountains Wildlife, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Qingfang Sun
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Jincan Li
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Yining Chen
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Yingjie Guo
- Biology Research Centre of Qin Mountains Wildlife, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
- College of Forestry, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Fangxia Yang
- Biology Research Centre of Qin Mountains Wildlife, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China.
- College of Forestry, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China.
| | - Wuzi Dong
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China.
- Biology Research Centre of Qin Mountains Wildlife, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China.
| |
Collapse
|
11
|
Gao X, Gao YY, Yan HY, Liu GJ, Zhou Y, Tao T, Yue TT, Pang C, Chen XX, Gao S, Wu LY, Hang CH, Li W. PDK4 Decrease Neuronal Apoptosis via Inhibiting ROS-ASK1/P38 Pathway in Early Brain Injury After Subarachnoid Hemorrhage. Antioxid Redox Signal 2022; 36:505-524. [PMID: 34498942 DOI: 10.1089/ars.2021.0083] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Aims: Metabolic disorders may play key roles in oxidative stress and neuronal apoptosis in response to early brain injury (EBI) after subarachnoid hemorrhage (SAH). Pyruvate dehydrogenase (PDH) is related to oxidative stress in EBI, and its activity obviously decreases after SAH. We discovered that only pyruvate dehydrogenase kinase 4 (PDK4) expression was obviously increased among the four PDK isozymes after SAH in preliminary experiments. Therefore, we attempted to investigate the effects and corresponding mechanisms of PDK4 on oxidative stress after SAH. Results: First, we confirmed that PDK4 overexpression promoted PDH phosphorylation, inhibited PDH activity, and changed cell metabolism after SAH. A small interfering RNA (siRNA) targeting PDK4, a lentiviral PDK4 overexpression vector, and dichloroacetic acid (DCA) were used to regulate the expression and activity of PDK4. The siRNA decreased PDH phosphorylation, promoted reactive oxygen species (ROS) production, activated the apoptosis signal-regulating kinase 1 (ASK1)/P38 pathway, and induced neuronal apoptosis. The lentivirus further attenuated PDH activity, oxidative stress, and neuronal apoptosis. DCA inhibited the activity of PDK4, but increased the expression of PDK4 due to a feedback mechanism. Inactivated PDK4 did not effectively suppress PDH activity, which increased ROS production, activated the ASK1/P38 pathway, and led to neuronal apoptosis. Innovation: This study provides new insights into the potential antioxidant and antiapoptotic effects of the PDK4-PDH axis on EBI after SAH. Conclusions: The early overexpression of PDK4 after SAH may attenuate neuronal apoptosis by reducing oxidative stress via the ROS/ASK1/P38 pathway. PDK4 may be a new potential therapeutic target to ameliorate EBI after SAH. Antioxid. Redox Signal. 36, 505-524.
Collapse
Affiliation(s)
- Xuan Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yong-Yue Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hui-Ying Yan
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Guang-Jie Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yan Zhou
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Tao Tao
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Ting-Ting Yue
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Cong Pang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiang-Xin Chen
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Sen Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ling-Yun Wu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
12
|
A “Weird” Mitochondrial Fatty Acid Oxidation as a Metabolic “Secret” of Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2339584. [PMID: 35178152 PMCID: PMC8847026 DOI: 10.1155/2022/2339584] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/29/2021] [Indexed: 12/15/2022]
Abstract
Cancer metabolism is an extensively studied field since the discovery of the Warburg effect about 100 years ago and continues to be increasingly intriguing and enigmatic so far. It has become clear that glycolysis is not the only abnormally activated metabolic pathway in the cancer cells, but the same is true for the fatty acid synthesis (FAS) and mevalonate pathway. In the last decade, a lot of data have been accumulated on the pronounced mitochondrial fatty acid oxidation (mFAO) in many types of cancer cells. In this article, we discuss how mFAO can escape normal regulation under certain conditions and be overactivated. Such abnormal activation of mitochondrial β-oxidation can also be combined with mutations in certain enzymes of the Krebs cycle that are common in cancer. If overactivated β-oxidation is combined with other common cancer conditions, such as dysfunctions in the electron transport complexes, and/or hypoxia, this may alter the redox state of the mitochondrial matrix. We propose the idea that the altered mitochondrial redox state and/or inhibited Krebs cycle at certain segments may link mitochondrial β-oxidation to the citrate-malate shuttle instead to the Krebs cycle. We call this abnormal metabolic condition “β-oxidation shuttle”. It is unconventional mFAO, a separate metabolic pathway, unexplored so far as a source of energy, as well as a source of cataplerosis, leading to biomass accumulation, accelerated oxygen consumption, and ultimately a source of proliferation. It is inefficient as an energy source and must consume significantly more oxygen per mole of ATP produced when combined with acetyl-CoA consuming pathways, such as the FAS and mevalonate pathway.
Collapse
|
13
|
Chen WM, Chen YM, Jiang SY, Tao YP, Hong YG, Yang L, Zheng H, Chen JQ. LncRNA POT1-AS1 accelerates the progression of gastric cancer by serving as a competing endogenous RNA of microRNA-497-5p to increase PDK3 expression. J Gastrointest Oncol 2021; 12:2728-2742. [PMID: 35070402 PMCID: PMC8748042 DOI: 10.21037/jgo-21-709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/25/2021] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is the most common malignant tumor of the digestive system. Although progress has been reported in terms of treatment, it is still the second leading cause of cancer-related death. Long non-coding RNAs have been shown to play a key role in human cancers in recent investigations. However, the role of POT1-AS1 in GC is still unclear. METHODS The relative POT1-AS1 level in GC tissues and paracancerous tissues was detected by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). The biological function of POT1-AS1 was studied by CCK8 and Transwell assays in vitro experiments. Moreover, the downstream target genes of POT1-AS1 were predicted by bioinformatics. RESULTS In this study, high POT1-AS1 expression in GC cells was confirmed, and its elevated expression was linked to patients' negative clinicopathological characteristics, as well as shorter disease-free survival (DFS) and overall survival (OS). POT1-AS1 was shown to serve as a competing endogenous RNA (ceRNA) by sponging miR-497-5p to increase PDK3 expression. The impact of POT1-AS1 silencing on GC malignant phenotypes could be reversed by suppressing miR-497-5p or restoring PDK3, according to rescue experiments. CONCLUSIONS In brief, POT1-AS1 acted as an oncogenic lncRNA in GC, facilitating GC development by affecting the miR-497-5p/PDK3 axis, implying that the POT1-AS1/miR-497-5p/PDK3 axis is a useful target in anticancer therapy.
Collapse
Affiliation(s)
- Wei-Min Chen
- Department of Gastroenterology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Yi-Ming Chen
- Department of Gastroenterology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Si-Yuan Jiang
- Department of Colorectal cancer, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuan-Ping Tao
- Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai, China
| | - Yong-Gang Hong
- Department of Colorectal cancer, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Le Yang
- Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai, China
| | - Hao Zheng
- Shanghai Key Laboratory of Hepatobiliary Tumor Biology (EHBH), Shanghai, China
- Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jian-Qing Chen
- Department of Gastroenterology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
14
|
Freitas-Lima LC, Budu A, Estrela GR, da Silva TA, Arruda AC, de Carvalho Araujo R. Metabolic fasting stress is ameliorated in Kinin B1 receptor-deficient mice. Life Sci 2021; 294:120007. [PMID: 34600938 DOI: 10.1016/j.lfs.2021.120007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
The liver has an essential role in responding to metabolic demands under stress conditions. The organ stores, releases, and recycles metabolism-related substrates. However, it is not clear how the Kallikrein-Kinin System modulates metabolic flexibility shift between energetic sources. AIMS To analyze the hepatic metabolism in kinin B1 receptor deficient mice (B1KO mice) under fasting conditions. MAIN METHODS WT and B1KO male mice were allocated in a calorimetric cage for 7 days and 48 h before the euthanasia, half of the animals of both groups were under fasting conditions. Biochemical parameters, ketone bodies (KB), and gene expression involving the liver energetic metabolism genes were evaluated. KEY FINDINGS Kinin B1 receptor (B1R) modulates the metabolic shift under fasting conditions, reducing the VO2 expenditure. A preference for carbohydrates as an energetic source is suggested, as the B1KO group did not display an increase in KB in the serum. Moreover, the B1KO animals displayed higher serum triglycerides concentration compared to WT fasting mice. Interestingly, the lack of B1R induces the increase expression of enzymes from the glycolysis and lipolysis pathways under the fed. However, under fasting, the enzymatic expression of gluconeogenesis, glyceroneogenesis, and ketogenesis of these pathways does not occur, suggesting an absence of the shift metabolism responsivity, and this condition is modulated by PDK4 under FOXO1 control. SIGNIFICANCE B1R has an important role in the hepatic glucose metabolism, which in turn influences the energetic metabolism, and in long-term outcomes, such as in the decrease in hepatic glycogen stores and in the enhancement of hepatic metabolism.
Collapse
Affiliation(s)
| | - Alexandre Budu
- Department of Biophysics, Federal University of São Paulo, 04039032 São Paulo, Brazil.
| | - Gabriel Rufino Estrela
- Department of Medicine, Discipline of Nephrology, Federal University of São Paulo, São Paulo, Brazil; Department of Clinical and Experimental Oncology, Discipline of Hematology and Hematotherapy, Federal University of São Paulo, 04037002 São Paulo, Brazil.
| | - Thais Alves da Silva
- Department of Biophysics, Federal University of São Paulo, 04039032 São Paulo, Brazil.
| | - Adriano Cleis Arruda
- Department of Medicine, Discipline of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Ronaldo de Carvalho Araujo
- Department of Biophysics, Federal University of São Paulo, 04039032 São Paulo, Brazil; Department of Medicine, Discipline of Nephrology, Federal University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
15
|
The Regulatory Roles of PPARs in Skeletal Muscle Fuel Metabolism and Inflammation: Impact of PPAR Agonism on Muscle in Chronic Disease, Contraction and Sepsis. Int J Mol Sci 2021; 22:ijms22189775. [PMID: 34575939 PMCID: PMC8465345 DOI: 10.3390/ijms22189775] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
The peroxisome proliferator-activated receptor (PPAR) family of transcription factors has been demonstrated to play critical roles in regulating fuel selection, energy expenditure and inflammation in skeletal muscle and other tissues. Activation of PPARs, through endogenous fatty acids and fatty acid metabolites or synthetic compounds, has been demonstrated to have lipid-lowering and anti-diabetic actions. This review will aim to provide a comprehensive overview of the functions of PPARs in energy homeostasis, with a focus on the impacts of PPAR agonism on muscle metabolism and function. The dysregulation of energy homeostasis in skeletal muscle is a frequent underlying characteristic of inflammation-related conditions such as sepsis. However, the potential benefits of PPAR agonism on skeletal muscle protein and fuel metabolism under these conditions remains under-investigated and is an area of research opportunity. Thus, the effects of PPARγ agonism on muscle inflammation and protein and carbohydrate metabolism will be highlighted, particularly with its potential relevance in sepsis-related metabolic dysfunction. The impact of PPARδ agonism on muscle mitochondrial function, substrate metabolism and contractile function will also be described.
Collapse
|
16
|
Thirunavukarasu S, Brown LAE, Chowdhary A, Jex N, Swoboda P, Greenwood JP, Plein S, Levelt E. Rationale and design of the randomised controlled cross-over trial: Cardiovascular effects of empaglifozin in diabetes mellitus. Diab Vasc Dis Res 2021; 18:14791641211021585. [PMID: 34182806 PMCID: PMC8481726 DOI: 10.1177/14791641211021585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is associated with an increased risk of cardiovascular (CV) disease. In patients with T2D and established CV disease, selective inhibitors of sodium-glucose cotransporter 2 (SGLT2) have been shown to decrease CV and all-cause mortality, and heart failure (HF) admissions. Utilising CV magnetic resonance imaging (CMR) and continuous glucose monitoring (CGM) by FreeStyle Libre Pro Sensor, we aim to explore the mechanisms of action which give Empagliflozin, an SGLT2 inhibitor, its beneficial CV effects and compare these to the effects of dipeptidyl peptidase-4 inhibitor Sitagliptin. METHODS This is a single centre, open-label, cross-over trial conducted at the Leeds Teaching Hospitals NHS Trust. Participants are randomised for the order of treatment and receive 3 months therapy with Empagliflozin, and 3 months therapy with Sitagliptin sequentially. Twenty-eight eligible T2D patients with established ischaemic heart disease will be recruited. Patients undergo serial CMR scans on three visits. DISCUSSION The primary outcome measure is the myocardial perfusion reserve in remote myocardium. We hypothesise that Empaglifozin treatment is associated with improvements in myocardial blood flow and reductions in myocardial interstitial fibrosis, independent of CGM measured glycemic control in patients with T2D and established CV disease. TRIAL REGISTRATION This study has full research ethics committee approval (REC: 18/YH/0190) and data collection is anticipated to finish in December 2021. This study was retrospectively registered at https://doi.org/10.1186/ISRCTN82391603 and monitored by the University of Leeds. The study results will be submitted for publication within 6 months of completion.
Collapse
Affiliation(s)
- Sharmaine Thirunavukarasu
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Louise AE Brown
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Amrit Chowdhary
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Nicholas Jex
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Peter Swoboda
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - John P Greenwood
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Sven Plein
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Eylem Levelt
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
17
|
Pyruvate dehydrogenase kinases (PDKs): an overview toward clinical applications. Biosci Rep 2021; 41:228121. [PMID: 33739396 PMCID: PMC8026821 DOI: 10.1042/bsr20204402] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 01/01/2023] Open
Abstract
Pyruvate dehydrogenase kinase (PDK) can regulate the catalytic activity of pyruvate decarboxylation oxidation via the mitochondrial pyruvate dehydrogenase complex, and it further links glycolysis with the tricarboxylic acid cycle and ATP generation. This review seeks to elucidate the regulation of PDK activity in different species, mainly mammals, and the role of PDK inhibitors in preventing increased blood glucose, reducing injury caused by myocardial ischemia, and inducing apoptosis of tumor cells. Regulations of PDKs expression or activity represent a very promising approach for treatment of metabolic diseases including diabetes, heart failure, and cancer. The future research and development could be more focused on the biochemical understanding of the diseases, which would help understand the cellular energy metabolism and its regulation by pharmacological effectors of PDKs.
Collapse
|
18
|
Dias IB, Bouma HR, Henning RH. Unraveling the Big Sleep: Molecular Aspects of Stem Cell Dormancy and Hibernation. Front Physiol 2021; 12:624950. [PMID: 33867999 PMCID: PMC8047423 DOI: 10.3389/fphys.2021.624950] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 03/11/2021] [Indexed: 12/14/2022] Open
Abstract
Tissue-resident stem cells may enter a dormant state, also known as quiescence, which allows them to withstand metabolic stress and unfavorable conditions. Similarly, hibernating mammals can also enter a state of dormancy used to evade hostile circumstances, such as food shortage and low ambient temperatures. In hibernation, the dormant state of the individual and its cells is commonly known as torpor, and is characterized by metabolic suppression in individual cells. Given that both conditions represent cell survival strategies, we here compare the molecular aspects of cellular quiescence, particularly of well-studied hematopoietic stem cells, and torpor at the cellular level. Critical processes of dormancy are reviewed, including the suppression of the cell cycle, changes in metabolic characteristics, and cellular mechanisms of dealing with damage. Key factors shared by hematopoietic stem cell quiescence and torpor include a reversible activation of factors inhibiting the cell cycle, a shift in metabolism from glucose to fatty acid oxidation, downregulation of mitochondrial activity, key changes in hypoxia-inducible factor one alpha (HIF-1α), mTOR, reversible protein phosphorylation and autophagy, and increased radiation resistance. This similarity is remarkable in view of the difference in cell populations, as stem cell quiescence regards proliferating cells, while torpor mainly involves terminally differentiated cells. A future perspective is provided how to advance our understanding of the crucial pathways that allow stem cells and hibernating animals to engage in their 'great slumbers.'
Collapse
Affiliation(s)
- Itamar B. Dias
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hjalmar R. Bouma
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Robert H. Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
19
|
Blocking Aerobic Glycolysis by Targeting Pyruvate Dehydrogenase Kinase in Combination with EGFR TKI and Ionizing Radiation Increases Therapeutic Effect in Non-Small Cell Lung Cancer Cells. Cancers (Basel) 2021; 13:cancers13050941. [PMID: 33668151 PMCID: PMC7956357 DOI: 10.3390/cancers13050941] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Non-small cell lung cancer (NSCLC) patients harboring oncogenic mutations in the epidermal growth factor receptor (EGFR) inevitably develop resistance to targeted EGFR tyrosine kinase inhibitors (TKI) therapy. To support malignant features associated with cancer development and therapy resistance, the cancer cells adapt their metabolic rate and pathways. As an example, aerobic glycolysis, where the cells use glycolysis in the presence of oxygen, is frequently seen. Here we show that targeting aerobic glycolysis represents a promising strategy in cancer therapeutics. Abstract Increased glycolytic activity is a hallmark of cancer initiation and progression and is often observed in non-small cell lung cancer (NSCLC). Pyruvate dehydrogenase (PDH) complex acts as a gatekeeper between glycolysis and oxidative phosphorylation, and activation of PDH is known to inhibit glycolytic activity. As part of a standard therapeutic regimen, patients with NSCLC harboring oncogenic mutations in the epidermal growth factor receptor (EGFR) are treated with EGFR tyrosine kinase inhibitors (EGFR TKIs). Independent of good initial response, development of resistance to this therapy is inevitable. In the presented work, we propose that inhibition of glycolysis will add to the therapeutic effects and possibly prevent development of resistance against both EGFR TKIs and ionizing radiation in NSCLC. Analysis of transcriptome data from two independent NSCLC patient cohorts identified increased expression of pyruvate dehydrogenase kinase 1 (PDHK1) as well as upregulated expression of genes involved in glucose metabolism in tumors compared to normal tissue. We established in vitro models of development of resistance to EGFR TKIs to study metabolism and determine if targeting PDHK would prevent development of resistance to EGFR TKIs in NSCLC cells. The PDHK1 inhibitor dichloroacetate (DCA) in combination with EGFR TKIs and/or ionizing radiation was shown to increase the therapeutic effect in our NSCLC cell models. This mechanism was associated with redirected metabolism towards pyruvate oxidation and reduced lactate production, both in EGFR TKI sensitive and resistant NSCLC cells. Using DCA, the intracellular pool of pyruvate available for lactic fermentation becomes limited. Consequently, pyruvate is redirected to the mitochondria, and reinforces mitochondrial activity. Addition of DCA to cell culture deacidifies the extracellular microenvironment as less lactate is produced and excreted. In our study, we find that this redirection of metabolism adds to the therapeutic effect of EGFR TKI and ionizing radiation in NSCLC.
Collapse
|
20
|
RNA-sequencing analysis reveals the potential contribution of lncRNAs in palmitic acid-induced insulin resistance of skeletal muscle cells. Biosci Rep 2020; 40:221488. [PMID: 31833538 PMCID: PMC6944669 DOI: 10.1042/bsr20192523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/28/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Insulin resistance (IR) has been considered as the common pathological basis and developmental driving force for most metabolic diseases. Long noncoding RNAs (lncRNAs) have emerged as pivotal regulators in modulation of glucose and lipid metabolism. However, the comprehensive profile of lncRNAs in skeletal muscle cells under the insulin resistant status and the possible biological effects of them were not fully studied. In this research, using C2C12 myotubes as cell models in vitro, deep RNA-sequencing was performed to profile lncRNAs and mRNAs between palmitic acid-induced IR C2C12 myotubes and control ones. The results revealed that a total of 144 lncRNAs including 70 up-regulated and 74 down-regulated (|fold change| > 2, q < 0.05) were significantly differentially expressed in palmitic acid-induced insulin resistant cells. In addition, functional annotation analysis based on the Gene Ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) databases revealed that the target genes of the differentially expressed lncRNAs were significantly enriched in fatty acid oxidation, lipid oxidation, PPAR signaling pathway, and insulin signaling pathway. Moreover, Via qPCR, most of selected lncRNAs in myotubes and db/db mice skeletal muscle showed the consistent expression trends with RNA-sequencing. Co-expression analysis also explicated the key lncRNA–mRNA interactions and pointed out a potential regulatory network of candidate lncRNA ENSMUST00000160839. In conclusion, the present study extended the skeletal muscle lncRNA database and provided novel potential regulators for future genetic and molecular studies on insulin resistance, which is helpful for prevention and treatment of the related metabolic diseases.
Collapse
|
21
|
Lin L, Zhang S, Lin Y, Liu W, Zou B, Cai Y, Liu D, Sun Y, Zhong Y, Xiao D, Liao Q, Xie Z. Untargeted metabolomics analysis on Cicer arietinium L.-Induced Amelioration in T2D rats by UPLC-Q-TOF-MS/MS. JOURNAL OF ETHNOPHARMACOLOGY 2020; 261:113013. [PMID: 32526338 DOI: 10.1016/j.jep.2020.113013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/24/2020] [Accepted: 05/22/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cicer arietinium L., which belongs to Cicer genus, was not only a kind of traditional Chinese medicines (TCM) recorded in Pharmacopoeia of the People's Republic of China (version 2015), but also a kind of Uighur antidiabetic medicines. It has been used as an adjuvant drug or functional food for thousand years in Xinjiang province, China. However, the mechanisms of C. arietinium treatment in T2D have not been fully understood especially on the perspective of metabolomics. AIM OF THE STUDY To clarify the potential mechanisms of C. arietinium treatment in T2D from the perspective of metabolomics since T2D is indeed a kind of metabolic syndromes. MATERIALS AND METHODS T2D rat model was built by HFD for 4 weeks, combining with STZ administration. T2D rats were administrated C. arietinium extraction or metformin (positive control) for 4 weeks. UPLC-Q-TOF-MS was applied to screen and identify differential metabolites among groups. RESULTS After 4 weeks of treatments, IR and inflammation were greatly ameliorated in C. arietinium group. And the therapeutic efficiency of C. arietinium treatment was comparable to metformin treatment. Differential metabolites related to C. arietinium treatment, including acylcarnitines, amino acid related metabolites and organic acids, were further used to indicate relevant pathways in T2D rats, including glyoxylate and dicarboxylate metabolism, tricarboxylic acid cycle, vitamin B6 metabolism and energy metabolism. CONCLUSIONS In summary, C. arietinium treatment could effectively alleviate diabetic symptoms and regulate metabolic disorders in T2D rats.
Collapse
MESH Headings
- Animals
- Biomarkers/blood
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Chromatography, High Pressure Liquid
- Cicer/chemistry
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/drug therapy
- Energy Metabolism/drug effects
- Hypoglycemic Agents/isolation & purification
- Hypoglycemic Agents/pharmacology
- Male
- Metabolomics
- Metformin/pharmacology
- Plant Extracts/isolation & purification
- Plant Extracts/pharmacology
- Rats, Sprague-Dawley
- Spectrometry, Mass, Electrospray Ionization
- Streptozocin
- Tandem Mass Spectrometry
Collapse
Affiliation(s)
- Lei Lin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shaobao Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yixuan Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wen Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510006, China
| | - Baorong Zou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510006, China
| | - Ying Cai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510006, China
| | - Deliang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510006, China
| | - Yangwen Sun
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuping Zhong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Dan Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510006, China
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, 510006, China.
| |
Collapse
|
22
|
Posa DK, Baba SP. Intracellular pH Regulation of Skeletal Muscle in the Milieu of Insulin Signaling. Nutrients 2020; 12:nu12102910. [PMID: 32977552 PMCID: PMC7598285 DOI: 10.3390/nu12102910] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 12/18/2022] Open
Abstract
Type 2 diabetes (T2D), along with obesity, is one of the leading health problems in the world which causes other systemic diseases, such as cardiovascular diseases and kidney failure. Impairments in glycemic control and insulin resistance plays a pivotal role in the development of diabetes and its complications. Since skeletal muscle constitutes a significant tissue mass of the body, insulin resistance within the muscle is considered to initiate the onset of diet-induced metabolic syndrome. Insulin resistance is associated with impaired glucose uptake, resulting from defective post-receptor insulin responses, decreased glucose transport, impaired glucose phosphorylation, oxidation and glycogen synthesis in the muscle. Although defects in the insulin signaling pathway have been widely studied, the effects of cellular mechanisms activated during metabolic syndrome that cross-talk with insulin responses are not fully elucidated. Numerous reports suggest that pathways such as inflammation, lipid peroxidation products, acidosis and autophagy could cross-talk with insulin-signaling pathway and contribute to diminished insulin responses. Here, we review and discuss the literature about the defects in glycolytic pathway, shift in glucose utilization toward anaerobic glycolysis and change in intracellular pH [pH]i within the skeletal muscle and their contribution towards insulin resistance. We will discuss whether the derangements in pathways, which maintain [pH]i within the skeletal muscle, such as transporters (monocarboxylate transporters 1 and 4) and depletion of intracellular buffers, such as histidyl dipeptides, could lead to decrease in [pH]i and the onset of insulin resistance. Further we will discuss, whether the changes in [pH]i within the skeletal muscle of patients with T2D, could enhance the formation of protein aggregates and activate autophagy. Understanding the mechanisms by which changes in the glycolytic pathway and [pH]i within the muscle, contribute to insulin resistance might help explain the onset of obesity-linked metabolic syndrome. Finally, we will conclude whether correcting the pathways which maintain [pH]i within the skeletal muscle could, in turn, be effective to maintain or restore insulin responses during metabolic syndrome.
Collapse
Affiliation(s)
- Dheeraj Kumar Posa
- Diabetes and Obesity Center, University of Louisville, Louisville, KY 40202, USA
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| | - Shahid P Baba
- Diabetes and Obesity Center, University of Louisville, Louisville, KY 40202, USA
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
23
|
Jeon JH, Thoudam T, Choi EJ, Kim MJ, Harris RA, Lee IK. Loss of metabolic flexibility as a result of overexpression of pyruvate dehydrogenase kinases in muscle, liver and the immune system: Therapeutic targets in metabolic diseases. J Diabetes Investig 2020; 12:21-31. [PMID: 32628351 PMCID: PMC7779278 DOI: 10.1111/jdi.13345] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Good health depends on the maintenance of metabolic flexibility, which in turn is dependent on the maintenance of regulatory flexibility of a large number of regulatory enzymes, but especially the pyruvate dehydrogenase complex (PDC), because of its central role in carbohydrate metabolism. Flexibility in regulation of PDC is dependent on rapid changes in the phosphorylation state of PDC determined by the relative activities of the pyruvate dehydrogenase kinases (PDKs) and the pyruvate dehydrogenase phosphatases. Inactivation of the PDC by overexpression of PDK4 contributes to hyperglycemia, and therefore the serious health problems associated with diabetes. Loss of regulatory flexibility of PDC occurs in other disease states and pathological conditions that have received less attention than diabetes. These include cancers, non‐alcoholic fatty liver disease, cancer‐induced cachexia, diabetes‐induced nephropathy, sepsis and amyotrophic lateral sclerosis. Overexpression of PDK4, and in some situations, the other PDKs, as well as under expression of the pyruvate dehydrogenase phosphatases, leads to inactivation of the PDC, mitochondrial dysfunction and deleterious effects with health consequences. The possible basis for this phenomenon, along with evidence that overexpression of PDK4 results in phosphorylation of “off‐target” proteins and promotes excessive transport of Ca2+ into mitochondria through mitochondria‐associated endoplasmic reticulum membranes are discussed. Recent efforts to find small molecule PDK inhibitors with therapeutic potential are also reviewed.
Collapse
Affiliation(s)
- Jae-Han Jeon
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea.,Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Korea
| | - Themis Thoudam
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Korea
| | - Eun Jung Choi
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Korea
| | - Min-Ji Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea
| | - Robert A Harris
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea.,Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Korea.,Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Korea.,Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Korea
| |
Collapse
|
24
|
Høgild ML, Gudiksen A, Pilegaard H, Stødkilde-Jørgensen H, Pedersen SB, Møller N, Jørgensen JOL, Jessen N. Redundancy in regulation of lipid accumulation in skeletal muscle during prolonged fasting in obese men. Physiol Rep 2020; 7:e14285. [PMID: 31724339 PMCID: PMC6854099 DOI: 10.14814/phy2.14285] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Fasting in human subjects shifts skeletal muscle metabolism toward lipid utilization and accumulation, including intramyocellular lipid (IMCL) deposition. Growth hormone (GH) secretion amplifies during fasting and promotes lipolysis and lipid oxidation, but it is unknown to which degree lipid deposition and metabolism in skeletal muscle during fasting depends on GH action. To test this, we studied nine obese but otherwise healthy men thrice: (a) in the postabsorptive state (“CTRL”), (b) during 72‐hr fasting (“FAST”), and (c) during 72‐hr fasting and treatment with a GH antagonist (GHA) (“FAST + GHA”). IMCL was assessed by magnetic resonance spectroscopy (MRS) and blood samples were drawn for plasma metabolomics assessment while muscle biopsies were obtained for measurements of regulators of substrate metabolism. Prolonged fasting was associated with elevated GH levels and a pronounced GHA‐independent increase in circulating medium‐ and long‐chain fatty acids, glycerol, and ketone bodies indicating increased supply of lipid intermediates to skeletal muscle. Additionally, fasting was associated with a release of short‐, medium‐, and long‐chain acylcarnitines to the circulation from an increased β‐oxidation. This was consistent with a ≈55%–60% decrease in pyruvate dehydrogenase (PDHa) activity. Opposite, IMCL content increased ≈75% with prolonged fasting without an effect of GHA. We suggest that prolonged fasting increases lipid uptake in skeletal muscle and saturates lipid oxidation, both favoring IMCL deposition. This occurs without a detectable effect of GHA on skeletal muscle lipid metabolism.
Collapse
Affiliation(s)
- Morten L Høgild
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anders Gudiksen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Hans Stødkilde-Jørgensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,The MR Research Center, Aarhus University Hospital, Copenhagen, Denmark
| | - Steen Bønløkke Pedersen
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Niels Møller
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens O L Jørgensen
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Niels Jessen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
25
|
Effect of Cod Residual Protein Supplementation on Markers of Glucose Regulation in Lean Adults: A Randomized Double-Blind Study. Nutrients 2020; 12:nu12051445. [PMID: 32429429 PMCID: PMC7285039 DOI: 10.3390/nu12051445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/05/2020] [Accepted: 05/13/2020] [Indexed: 12/18/2022] Open
Abstract
Large quantities of protein-rich cod residuals, which are currently discarded, could be utilized for human consumption. Although fish fillet intake is related to beneficial health effects, little is known about the potential health effects of consuming cod residual protein powder. Fifty lean adults were randomized to consume capsules with 8.1 g/day of cod residual protein (Cod-RP) or placebo capsules (Control group) for eight weeks, in this randomized, double-blind study. The intervention was completed by 40 participants. Fasting glucose and insulin concentrations were unaffected by Cod-RP supplementation, whereas plasma concentrations of α-hydroxybutyrate, β-hydroxybutyrate and acetoacetate all were decreased compared with the Control group. Trimethylamine N-oxide concentration in plasma and urine were increased in the Cod-RP group compared with the Control group. To conclude, the reduction in these potential early markers of impaired glucose metabolism following Cod-RP supplementation may indicate beneficial glucoregulatory effects of cod residual proteins. Trimethylamine N-oxide appears to be an appropriate biomarker of cod residual protein intake in lean adults.
Collapse
|
26
|
Metformin and trimetazidine ameliorate diabetes-induced cognitive impediment in status epileptic rats. Epilepsy Behav 2020; 104:106893. [PMID: 32000097 DOI: 10.1016/j.yebeh.2019.106893] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/24/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023]
Abstract
Patients with diabetes and epilepsy are more prone to cognitive impairment, dementia, and even Alzheimer's disease. Diabetes-induced inflammatory process is one of the main contributing factors; however, the impact on seizure is not clear. The current study is aimed to examine the role of metformin and trimetazidine in the reduction of neuronal damage caused by inflammatory mediators and apoptotic factors in diabetic epileptic rodent model. Diabetic epileptic rats received orally either metformin (100 mg/kg) or trimetazidine (10 mg/kg) for 3 weeks exhibited reduced cognitive function and ameliorated the disturbed brain neurotransmission. Besides, they improved both the inflammatory status and the histopathologic alterations. Administration of metformin or trimetazidine ameliorated the deterioration in cognitive function in Morris water maze (MWM) and reduced seizure score. Furthermore, brain neurotransmitters glutamate and γ-aminobutyric acid (GABA) were reverted back to their normal values. Both treatments reduced the rise in inflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α), apoptotic markers nuclear factor-κB (NF-κB) and caspase-3, and improved the pathological photomicrograph of the hippocampus of diabetic epileptic rats. Such effects were closely correlated to the observed increase in the adenosine triphosphate and adenosine diphosphate (ATP/ADP) ratio and reduction of death-associated protein (DAP) and mammalian target of rapamycin (mTOR). In conclusion, the current study shed light on the potential neuroprotective role of metformin and trimetazidine in the amelioration of cognitive function via hindering inflammatory processes in diabetic epileptic rats.
Collapse
|
27
|
Athithan L, Gulsin GS, McCann GP, Levelt E. Diabetic cardiomyopathy: Pathophysiology, theories and evidence to date. World J Diabetes 2019; 10:490-510. [PMID: 31641426 PMCID: PMC6801309 DOI: 10.4239/wjd.v10.i10.490] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 02/05/2023] Open
Abstract
The prevalence of type 2 diabetes (T2D) has increased worldwide and doubled over the last two decades. It features among the top 10 causes of mortality and morbidity in the world. Cardiovascular disease is the leading cause of complications in diabetes and within this, heart failure has been shown to be the leading cause of emergency admissions in the United Kingdom. There are many hypotheses and well-evidenced mechanisms by which diabetic cardiomyopathy as an entity develops. This review aims to give an overview of these mechanisms, with particular emphasis on metabolic inflexibility. T2D is associated with inefficient substrate utilisation, an inability to increase glucose metabolism and dependence on fatty acid oxidation within the diabetic heart resulting in mitochondrial uncoupling, glucotoxicity, lipotoxicity and initially subclinical cardiac dysfunction and finally in overt heart failure. The review also gives a concise update on developments within clinical imaging, specifically cardiac magnetic resonance studies to characterise and phenotype early cardiac dysfunction in T2D. A better understanding of the pathophysiology involved provides a platform for targeted therapy in diabetes to prevent the development of early heart failure with preserved ejection fraction.
Collapse
Affiliation(s)
- Lavanya Athithan
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Gaurav S Gulsin
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Gerald P McCann
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Eylem Levelt
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LF9 7TF, United Kingdom
| |
Collapse
|
28
|
Pendleton AL, Humphreys LR, Davis MA, Camacho LE, Anderson MJ, Limesand SW. Increased pyruvate dehydrogenase activity in skeletal muscle of growth-restricted ovine fetuses. Am J Physiol Regul Integr Comp Physiol 2019; 317:R513-R520. [PMID: 31314546 PMCID: PMC6842904 DOI: 10.1152/ajpregu.00106.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/16/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022]
Abstract
Fetal sheep with placental insufficiency-induced intrauterine growth restriction (IUGR) have lower fractional rates of glucose oxidation and greater gluconeogenesis, indicating lactate shuttling between skeletal muscle and liver. Suppression of pyruvate dehydrogenase (PDH) activity was proposed because of greater pyruvate dehydrogenase kinase (PDK) 4 and PDK1 mRNA concentrations in IUGR muscle. Although PDK1 and PDK4 inhibit PDH activity to reduce pyruvate metabolism, PDH protein concentrations and activity have not been examined in skeletal muscle from IUGR fetuses. Therefore, we evaluated the protein concentrations and activity of PDH and the kinases and phosphatases that regulate PDH phosphorylation status in the semitendinosus muscle from placenta insufficiency-induced IUGR sheep fetuses and control fetuses. Immunoblots were performed for PDH, phosphorylated PDH (E1α), PDK1, PDK4, and pyruvate dehydrogenase phosphatase 1 and 2 (PDP1 and PDP2, respectively). Additionally, the PDH, lactate dehydrogenase (LDH), and citrate synthase (CS) enzymatic activities were measured. Phosphorylated PDH concentrations were 28% lower (P < 0.01) and PDH activity was 67% greater (P < 0.01) in IUGR fetal muscle compared with control. PDK1, PDK4, PDP1, PDP2, and PDH concentrations were not different between groups. CS and LDH activities were also unaffected. Contrary to the previous speculation, PDH activity was greater in skeletal muscle from IUGR fetuses, which parallels lower phosphorylated PDH. Therefore, greater expression of PDK1 and PDK4 mRNA did not translate to greater PDK1 or PDK4 protein concentrations or inhibition of PDH as proposed. Instead, these findings show greater PDH activity in IUGR fetal muscle, which indicates that alternative regulatory mechanisms are responsible for lower pyruvate catabolism.
Collapse
Affiliation(s)
- Alexander L Pendleton
- Physiological Sciences Graduate Interdisciplinary Program, The University of Arizona, Tucson, Arizona
| | - Laurel R Humphreys
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona
| | - Melissa A Davis
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona
| | - Leticia E Camacho
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona
| | - Miranda J Anderson
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona
| | - Sean W Limesand
- Physiological Sciences Graduate Interdisciplinary Program, The University of Arizona, Tucson, Arizona
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona
| |
Collapse
|
29
|
Woolbright BL, Rajendran G, Harris RA, Taylor JA. Metabolic Flexibility in Cancer: Targeting the Pyruvate Dehydrogenase Kinase:Pyruvate Dehydrogenase Axis. Mol Cancer Ther 2019; 18:1673-1681. [PMID: 31511353 DOI: 10.1158/1535-7163.mct-19-0079] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/23/2019] [Accepted: 07/24/2019] [Indexed: 11/16/2022]
Abstract
Cancer cells use alterations of normal metabolic processes to sustain proliferation indefinitely. Transcriptional and posttranscriptional control of the pyruvate dehydrogenase kinase (PDK) family is one way in which cancer cells alter normal pyruvate metabolism to fuel proliferation. PDKs can phosphorylate and inactivate the pyruvate dehydrogenase complex (PDHC), which blocks oxidative metabolism of pyruvate by the mitochondria. This process is thought to enhance cancer cell growth by promoting anabolic pathways. Inhibition of PDKs induces cell death through increased PDH activity and subsequent increases in ROS production. The use of PDK inhibitors has seen widespread success as a potential therapeutic in laboratory models of multiple cancers; however, gaps still exist in our understanding of the biology of PDK regulation and function, especially in the context of individual PDKs. Efforts are currently underway to generate PDK-specific inhibitors and delineate the roles of individual PDK isozymes in specific cancers. The goal of this review is to understand the regulation of the PDK isozyme family, their role in cancer proliferation, and how to target this pathway therapeutically to specifically and effectively reduce cancer growth.
Collapse
Affiliation(s)
| | | | - Robert A Harris
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - John A Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
30
|
miR-497-5p inhibits gastric cancer cell proliferation and growth through targeting PDK3. Biosci Rep 2019; 39:BSR20190654. [PMID: 31409724 PMCID: PMC6732365 DOI: 10.1042/bsr20190654] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 06/30/2019] [Accepted: 08/09/2019] [Indexed: 01/17/2023] Open
Abstract
MicroRNA plays an important role in gastric cancer (GC) development, while the function of miR-497-5p in this disease remains unknown. In the present study, we demonstrated miR-497-5p as a tumor suppressive microRNA in GC. miR-497-5p was down-regulated in GC tissues and its expression was associated with the disease stage. Inhibition of miR-497-5p promoted GC cell proliferation and growth. By contrast, miR-497-5p ectopic expression suppressed the proliferation and growth of GC cells. In addition, miR-497-5p inhibited DNA synthesis and enhanced apoptosis in GC cells. The cell cycle progression was suppressed by miR-497-5p. Mechanistically, miR-497-5p directly targeted and suppressed the expression of pyruvate dehydrogenase kinase 3 (PDK3), which is highly expressed in GC tissues. Over-expression of PDK3 promoted the proliferation of GC cells. Our study revealed that miR-497-5p inhibited GC cell proliferation and growth via targeting PDK3.
Collapse
|
31
|
Pettersen IKN, Tusubira D, Ashrafi H, Dyrstad SE, Hansen L, Liu XZ, Nilsson LIH, Løvsletten NG, Berge K, Wergedahl H, Bjørndal B, Fluge Ø, Bruland O, Rustan AC, Halberg N, Røsland GV, Berge RK, Tronstad KJ. Upregulated PDK4 expression is a sensitive marker of increased fatty acid oxidation. Mitochondrion 2019; 49:97-110. [PMID: 31351920 DOI: 10.1016/j.mito.2019.07.009] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/01/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022]
Abstract
Fatty acid oxidation is a central fueling pathway for mitochondrial ATP production. Regulation occurs through multiple nutrient- and energy-sensitive molecular mechanisms. We explored if upregulated mRNA expression of the mitochondrial enzyme pyruvate dehydrogenase kinase 4 (PDK4) may be used as a surrogate marker of increased mitochondrial fatty acid oxidation, by indicating an overall shift from glucose to fatty acids as the preferred oxidation fuel. The association between fatty acid oxidation and PDK4 expression was studied in different contexts of metabolic adaption. In rats treated with the modified fatty acid tetradecylthioacetic acid (TTA), Pdk4 was upregulated simultaneously with fatty acid oxidation genes in liver and heart, whereas muscle and white adipose tissue remained unaffected. In MDA-MB-231 cells, fatty acid oxidation increased nearly three-fold upon peroxisome proliferator-activated receptor α (PPARα, PPARA) overexpression, and four-fold upon TTA-treatment. PDK4 expression was highly increased under these conditions. Further, overexpression of PDK4 caused increased fatty acid oxidation in these cells. Pharmacological activators of PPARα and AMPK had minor effects, while the mTOR inhibitor rapamycin potentiated the effect of TTA. There were minor changes in mitochondrial respiration, glycolytic function, and mitochondrial biogenesis under conditions of increased fatty acid oxidation. TTA was found to act as a mild uncoupler, which is likely to contribute to the metabolic effects. Repeated experiments with HeLa cells supported these findings. In summary, PDK4 upregulation implies an overarching metabolic shift towards increased utilization of fatty acids as energy fuel, and thus constitutes a sensitive marker of enhanced fatty acid oxidation.
Collapse
Affiliation(s)
| | | | - Hanan Ashrafi
- Department of Biomedicine, University of Bergen, Norway
| | | | - Lena Hansen
- Department of Biomedicine, University of Bergen, Norway; Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | | | | | | | | | - Hege Wergedahl
- Department of Sport, Food and Natural Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Bodil Bjørndal
- Department of Clinical Science, University of Bergen, Norway
| | - Øystein Fluge
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Ove Bruland
- Department of Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | | | - Nils Halberg
- Department of Biomedicine, University of Bergen, Norway
| | - Gro Vatne Røsland
- Department of Biomedicine, University of Bergen, Norway; Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Rolf Kristian Berge
- Department of Clinical Science, University of Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | | |
Collapse
|
32
|
Sharma A, Oonthonpan L, Sheldon RD, Rauckhorst AJ, Zhu Z, Tompkins SC, Cho K, Grzesik WJ, Gray LR, Scerbo DA, Pewa AD, Cushing EM, Dyle MC, Cox JE, Adams C, Davies BS, Shields RK, Norris AW, Patti G, Zingman LV, Taylor EB. Impaired skeletal muscle mitochondrial pyruvate uptake rewires glucose metabolism to drive whole-body leanness. eLife 2019; 8:e45873. [PMID: 31305240 PMCID: PMC6684275 DOI: 10.7554/elife.45873] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/15/2019] [Indexed: 12/13/2022] Open
Abstract
Metabolic cycles are a fundamental element of cellular and organismal function. Among the most critical in higher organisms is the Cori Cycle, the systemic cycling between lactate and glucose. Here, skeletal muscle-specific Mitochondrial Pyruvate Carrier (MPC) deletion in mice diverted pyruvate into circulating lactate. This switch disinhibited muscle fatty acid oxidation and drove Cori Cycling that contributed to increased energy expenditure. Loss of muscle MPC activity led to strikingly decreased adiposity with complete muscle mass and strength retention. Notably, despite decreasing muscle glucose oxidation, muscle MPC disruption increased muscle glucose uptake and whole-body insulin sensitivity. Furthermore, chronic and acute muscle MPC deletion accelerated fat mass loss on a normal diet after high fat diet-induced obesity. Our results illuminate the role of the skeletal muscle MPC as a whole-body carbon flux control point. They highlight the potential utility of modulating muscle pyruvate utilization to ameliorate obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Arpit Sharma
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Lalita Oonthonpan
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Ryan D Sheldon
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Adam J Rauckhorst
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Zhiyong Zhu
- Department of Internal Medicine, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Sean C Tompkins
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Kevin Cho
- Department of Chemistry, School of MedicineWashington UniversitySt. LouisUnited States
| | - Wojciech J Grzesik
- Fraternal Order of the Eagles Diabetes Research Center (FOEDRC), Carver College of MedicineUniversity of IowaIowa CityUnited States
- FOEDRC Metabolic Phenotyping Core Facility, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Lawrence R Gray
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Diego A Scerbo
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Alvin D Pewa
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Emily M Cushing
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Michael C Dyle
- Department of Internal Medicine, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - James E Cox
- Department of Biochemistry, School of MedicineUniversity of UtahSalt Lake CityUnited States
- Metabolomics Core Research Facility, School of MedicineUniversity of UtahSalt Lake CityUnited States
| | - Chris Adams
- Department of Internal Medicine, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Fraternal Order of the Eagles Diabetes Research Center (FOEDRC), Carver College of MedicineUniversity of IowaIowa CityUnited States
- Department of Molecular Physiology and Biophysics, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Pappajohn Biomedical Institute, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Abboud Cardiovascular Research Center, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Brandon S Davies
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Fraternal Order of the Eagles Diabetes Research Center (FOEDRC), Carver College of MedicineUniversity of IowaIowa CityUnited States
- Pappajohn Biomedical Institute, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Abboud Cardiovascular Research Center, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Richard K Shields
- Fraternal Order of the Eagles Diabetes Research Center (FOEDRC), Carver College of MedicineUniversity of IowaIowa CityUnited States
- Department of Physical Therapy and Rehabilitation Science, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Andrew W Norris
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Fraternal Order of the Eagles Diabetes Research Center (FOEDRC), Carver College of MedicineUniversity of IowaIowa CityUnited States
- FOEDRC Metabolic Phenotyping Core Facility, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Department of Pediatrics, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Gary Patti
- Department of Chemistry, School of MedicineWashington UniversitySt. LouisUnited States
| | - Leonid V Zingman
- Department of Internal Medicine, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Fraternal Order of the Eagles Diabetes Research Center (FOEDRC), Carver College of MedicineUniversity of IowaIowa CityUnited States
- Abboud Cardiovascular Research Center, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Department of Veterans Affairs, Medical Center, Carver College of MedicineUniversity of IowaIowa CityUnited States
| | - Eric B Taylor
- Department of Biochemistry, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Fraternal Order of the Eagles Diabetes Research Center (FOEDRC), Carver College of MedicineUniversity of IowaIowa CityUnited States
- Department of Molecular Physiology and Biophysics, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Pappajohn Biomedical Institute, Carver College of MedicineUniversity of IowaIowa CityUnited States
- Abboud Cardiovascular Research Center, Carver College of MedicineUniversity of IowaIowa CityUnited States
- FOEDRC Metabolomics Core Facility, Carver College of MedicineUniversity of IowaIowa CityUnited States
| |
Collapse
|
33
|
Pin F, Novinger LJ, Huot JR, Harris RA, Couch ME, O'Connell TM, Bonetto A. PDK4 drives metabolic alterations and muscle atrophy in cancer cachexia. FASEB J 2019; 33:7778-7790. [PMID: 30894018 DOI: 10.1096/fj.201802799r] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cachexia is frequently accompanied by severe metabolic derangements, although the mechanisms responsible for this debilitating condition remain unclear. Pyruvate dehydrogenase kinase (PDK)4, a critical regulator of cellular energetic metabolism, was found elevated in experimental models of cancer, starvation, diabetes, and sepsis. Here we aimed to investigate the link between PDK4 and the changes in muscle size in cancer cachexia. High PDK4 and abnormal energetic metabolism were found in the skeletal muscle of colon-26 tumor hosts, as well as in mice fed a diet enriched in Pirinixic acid, previously shown to increase PDK4 levels. Viral-mediated PDK4 overexpression in myotube cultures was sufficient to promote myofiber shrinkage, consistent with enhanced protein catabolism and mitochondrial abnormalities. On the contrary, blockade of PDK4 was sufficient to restore myotube size in C2C12 cultures exposed to tumor media. Our data support, for the first time, a direct role for PDK4 in promoting cancer-associated muscle metabolic alterations and skeletal muscle atrophy.-Pin, F., Novinger, L. J., Huot, J. R., Harris, R. A., Couch, M. E., O'Connell, T. M., Bonetto, A. PDK4 drives metabolic alterations and muscle atrophy in cancer cachexia.
Collapse
Affiliation(s)
- Fabrizio Pin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana University-Purdue University Indianapolis Center for Cachexia Research, Innovation, and Therapy, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Leah J Novinger
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Joshua R Huot
- Indiana University-Purdue University Indianapolis Center for Cachexia Research, Innovation, and Therapy, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert A Harris
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Marion E Couch
- Indiana University-Purdue University Indianapolis Center for Cachexia Research, Innovation, and Therapy, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Thomas M O'Connell
- Indiana University-Purdue University Indianapolis Center for Cachexia Research, Innovation, and Therapy, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrea Bonetto
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana University-Purdue University Indianapolis Center for Cachexia Research, Innovation, and Therapy, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
34
|
Kasper JD, Meyer RA, Beard DA, Wiseman RW. Effects of altered pyruvate dehydrogenase activity on contracting skeletal muscle bioenergetics. Am J Physiol Regul Integr Comp Physiol 2018; 316:R76-R86. [PMID: 30462525 DOI: 10.1152/ajpregu.00321.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
During aerobic exercise (>65% of maximum oxygen consumption), the primary source of acetyl-CoA to fuel oxidative ATP synthesis in muscle is the pyruvate dehydrogenase (PDH) reaction. This study investigated how regulation of PDH activity affects muscle energetics by determining whether activation of PDH with dichloroacetate (DCA) alters the dynamics of the phosphate potential of rat gastrocnemius muscle during contraction. Twitch contractions were induced in vivo over a broad range of intensities to sample submaximal and maximal aerobic workloads. Muscle phosphorus metabolites were measured in vivo before and after DCA treatment by phosphorus nuclear magnetic resonance spectroscopy. At rest, DCA increased PDH activation compared with control (90 ± 12% vs. 23 ± 3%, P < 0.05), with parallel decreases in inorganic phosphate (Pi) of 17% (1.4 ± 0.2 vs. 1.7 ± 0.1 mM, P < 0.05) and an increase in the free energy of ATP hydrolysis (ΔGATP) (-66.2 ± 0.3 vs. -65.6 ± 0.2 kJ/mol, P < 0.05). During stimulation DCA increased steady-state phosphocreatine (PCr) and the magnitude of ΔGATP, with concomitant reduction in Pi and ADP concentrations. These effects were not due to kinetic alterations in PCr hydrolysis, resynthesis, or glycolytic ATP production and altered the flow-force relationship between mitochondrial ATP synthesis rate and ΔGATP. DCA had no significant effect at 1.0- to 2.0-Hz stimulation because physiological mechanisms at these high stimulation levels cause maximal activation of PDH. These data support a role of PDH activation in the regulation of the energetic steady state by altering the phosphate potential (ΔGATP) at rest and during contraction.
Collapse
Affiliation(s)
- Jonathan D Kasper
- Department of Physiology, Michigan State University , East Lansing, Michigan
| | - Ronald A Meyer
- Department of Physiology, Michigan State University , East Lansing, Michigan.,Department of Radiology, Michigan State University , East Lansing, Michigan
| | - Daniel A Beard
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
| | - Robert W Wiseman
- Department of Physiology, Michigan State University , East Lansing, Michigan.,Department of Radiology, Michigan State University , East Lansing, Michigan
| |
Collapse
|
35
|
Svensson K, Dent JR, Tahvilian S, Martins VF, Sathe A, Ochala J, Patel MS, Schenk S. Defining the contribution of skeletal muscle pyruvate dehydrogenase α1 to exercise performance and insulin action. Am J Physiol Endocrinol Metab 2018; 315:E1034-E1045. [PMID: 30153068 PMCID: PMC6293170 DOI: 10.1152/ajpendo.00241.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The pyruvate dehydrogenase complex (PDC) converts pyruvate to acetyl-CoA and is an important control point for carbohydrate (CHO) oxidation. However, the importance of the PDC and CHO oxidation to muscle metabolism and exercise performance, particularly during prolonged or high-intensity exercise, has not been fully defined especially in mature skeletal muscle. To this end, we determined whether skeletal muscle-specific loss of pyruvate dehydrogenase alpha 1 ( Pdha1), which is a critical subunit of the PDC, impacts resting energy metabolism, exercise performance, or metabolic adaptation to high-fat diet (HFD) feeding. For this, we generated a tamoxifen (TMX)-inducible Pdha1 knockout (PDHmKO) mouse, in which PDC activity is temporally and specifically ablated in adult skeletal muscle. We assessed energy expenditure, ex vivo muscle contractile performance, and endurance exercise capacity in PDHmKO mice and wild-type (WT) littermates. Additionally, we studied glucose homeostasis and insulin sensitivity in muscle after 12 wk of HFD feeding. TMX administration largely ablated PDHα in skeletal muscle of adult PDHmKO mice but did not impact energy expenditure, muscle contractile function, or low-intensity exercise performance. Additionally, there were no differences in muscle insulin sensitivity or body composition in PDHmKO mice fed a control or HFD, as compared with WT mice. However, exercise capacity during high-intensity exercise was severely impaired in PDHmKO mice, in parallel with a large increase in plasma lactate concentration. In conclusion, although skeletal muscle PDC is not a major contributor to resting energy expenditure or long-duration, low-intensity exercise performance, it is necessary for optimal performance during high-intensity exercise.
Collapse
Affiliation(s)
- Kristoffer Svensson
- Department of Orthopaedic Surgery, University of California San Diego , La Jolla, California
| | - Jessica R Dent
- Department of Orthopaedic Surgery, University of California San Diego , La Jolla, California
| | - Shahriar Tahvilian
- Department of Orthopaedic Surgery, University of California San Diego , La Jolla, California
| | - Vitor F Martins
- Department of Orthopaedic Surgery, University of California San Diego , La Jolla, California
| | - Abha Sathe
- Department of Orthopaedic Surgery, University of California San Diego , La Jolla, California
| | - Julien Ochala
- School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London , London , United Kingdom
| | - Mulchand S Patel
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo, New York
| | - Simon Schenk
- Department of Orthopaedic Surgery, University of California San Diego , La Jolla, California
- Biomedical Sciences Graduate Program, University of California San Diego , La Jolla, California
| |
Collapse
|
36
|
Noguchi S, Kondo Y, Ito R, Katayama T, Kazama S, Kadota Y, Kitaura Y, Harris RA, Shimomura Y. Ca2+-dependent inhibition of branched-chain α-ketoacid dehydrogenase kinase by thiamine pyrophosphate. Biochem Biophys Res Commun 2018; 504:916-920. [DOI: 10.1016/j.bbrc.2018.09.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 09/07/2018] [Indexed: 11/26/2022]
|
37
|
Gudiksen A, Bertholdt L, Stankiewicz T, Villesen I, Bangsbo J, Plomgaard P, Pilegaard H. Training state and fasting-induced PDH regulation in human skeletal muscle. Pflugers Arch 2018; 470:1633-1645. [DOI: 10.1007/s00424-018-2164-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/17/2018] [Accepted: 06/07/2018] [Indexed: 12/13/2022]
|
38
|
Wu CY, Tso SC, Chuang JL, Gui WJ, Lou M, Sharma G, Khemtong C, Qi X, Wynn RM, Chuang DT. Targeting hepatic pyruvate dehydrogenase kinases restores insulin signaling and mitigates ChREBP-mediated lipogenesis in diet-induced obese mice. Mol Metab 2018; 12:12-24. [PMID: 29656110 PMCID: PMC6001905 DOI: 10.1016/j.molmet.2018.03.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 11/08/2022] Open
Abstract
Objective Mitochondrial pyruvate dehydrogenase kinases 1–4 (PDKs1–4) negatively regulate activity of the pyruvate dehydrogenase complex (PDC) by reversible phosphorylation. PDKs play a pivotal role in maintaining energy homeostasis and contribute to metabolic flexibility by attenuating PDC activity in various mammalian tissues. Cumulative evidence has shown that the up-regulation of PDK4 expression is tightly associated with obesity and diabetes. In this investigation, we test the central hypothesis that PDKs1-4 are a pharmacological target for lowering glucose levels and restoring insulin sensitivity in obesity and type 2 diabetes (T2D). Methods Diet-induced obese (DIO) mice were treated with a liver-specific pan-PDK inhibitor 2-[(2,4-dihydroxyphenyl) sulfonyl]isoindoline-4,6-diol (PS10) for four weeks, and results compared with PDK2/PDK4 double knockout (DKO) mice on the same high fat diet (HFD). Results Both PS10-treated DIO mice and HFD-fed DKO mice showed significantly improved glucose, insulin and pyruvate tolerance, compared to DIO controls, with lower plasma insulin levels and increased insulin signaling in liver. In response to lower glucose levels, phosphorylated AMPK in PS10-treated DIO and HFD-fed DKO mice is upregulated, accompanied by decreased nuclear carbohydrate-responsive element binding protein (ChREBP). The reduced ChREBP signaling correlates with down-regulation of hepatic lipogenic enzymes (ACC1, FAS, and SCD1), leading to markedly diminished hepatic steatosis in both study groups, with lower circulating cholesterol and triacylglyceride levels as well as reduced fat mass. PS10-treated DIO as well as DKO mice showed predominant fatty acid over glucose oxidation. However, unlike systemic DKO mice, increased hepatic PDC activity alone in PS10-treated DIO mice does not raise the plasma total ketone body level. Conclusion Our findings establish that specific targeting of hepatic PDKs with the PDK inhibitor PS10 is an effective therapeutic approach to maintaining glucose and lipid homeostasis in obesity and T2D, without the harmful ketoacidosis associated with systemic inhibition of PDKs. Diet-induced obese (DIO) mice were treated with a novel liver-specific pyruvate dehydrogenase kinase (PDK) inhibitor PS10. PS10-treated DIO mice and PDK2/PDK4-DKO mice fed indentical HFD showed improved glucose, insulin and pyruvate tolerance. PS10-treated DIO and untreated DKO mice manifest reduced hepatic steatosis and attenuation of ChREBP-mediated lipogenesis. Unlike DKO mice, targeting hepatic PDK alone in DIO mice with PS10 does not increase total plasma ketone-body levels.
Collapse
Affiliation(s)
- Cheng-Yang Wu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shih-Chia Tso
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jacinta L Chuang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wen-Jun Gui
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mingliang Lou
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Gaurav Sharma
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chalermchai Khemtong
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiangbing Qi
- Chemistry Center, National Institute of Biological Science, Beijing, China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - R Max Wynn
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - David T Chuang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
39
|
Gudiksen A, Pilegaard H. PGC-1 α and fasting-induced PDH regulation in mouse skeletal muscle. Physiol Rep 2017; 5:5/7/e13222. [PMID: 28400503 PMCID: PMC5392513 DOI: 10.14814/phy2.13222] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 02/04/2023] Open
Abstract
The purpose of the present study was to examine whether lack of skeletal muscle peroxisome proliferator‐activated receptor gamma coactivator 1 alpha (PGC‐1α) affects the switch in substrate utilization from a fed to fasted state and the fasting‐induced pyruvate dehydrogenase (PDH) regulation in skeletal muscle. Skeletal muscle‐specific PGC‐1α knockout (MKO) mice and floxed littermate controls were fed or fasted for 24 h. Fasting reduced PDHa activity, increased phosphorylation of all four known sites on PDH‐E1α and increased pyruvate dehydrogenase kinase (PDK4) and sirtuin 3 (SIRT3) protein levels, but did not alter total acetylation of PDH‐E1α. Lack of muscle PGC‐1α did not affect the switch from glucose to fat oxidation in the transition from the fed to fasted state, but was associated with lower and higher respiratory exchange ratio (RER) in the fed and fasted state, respectively. PGC‐1α MKO mice had lower skeletal muscle PDH‐E1α, PDK1, 2, 4, and pyruvate dehydrogenase phosphatase (PDP1) protein content than controls, but this did not prevent the fasting‐induced increase in PDH‐E1α phosphorylation in PGC‐1α MKO mice. However, lack of skeletal muscle PGC‐1α reduced SIRT3 protein content, increased total lysine PDH‐E1α acetylation in the fed state, and prevented a fasting‐induced increase in SIRT3 protein. In conclusion, skeletal muscle PGC‐1α is required for fasting‐induced upregulation of skeletal muscle SIRT3 and maintaining high fat oxidation in the fasted state, but is dispensable for preserving the capability to switch substrate during the transition from the fed to the fasted state and for fasting‐induced PDH regulation in skeletal muscle.
Collapse
Affiliation(s)
- Anders Gudiksen
- Section for Cell Biology and Physiology, August Krogh Building, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Pilegaard
- Section for Cell Biology and Physiology, August Krogh Building, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Desseille C, Deforges S, Biondi O, Houdebine L, D'amico D, Lamazière A, Caradeuc C, Bertho G, Bruneteau G, Weill L, Bastin J, Djouadi F, Salachas F, Lopes P, Chanoine C, Massaad C, Charbonnier F. Specific Physical Exercise Improves Energetic Metabolism in the Skeletal Muscle of Amyotrophic-Lateral- Sclerosis Mice. Front Mol Neurosci 2017; 10:332. [PMID: 29104532 PMCID: PMC5655117 DOI: 10.3389/fnmol.2017.00332] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/02/2017] [Indexed: 01/22/2023] Open
Abstract
Amyotrophic Lateral Sclerosis is an adult-onset neurodegenerative disease characterized by the specific loss of motor neurons, leading to muscle paralysis and death. Although the cellular mechanisms underlying amyotrophic lateral sclerosis (ALS)-induced toxicity for motor neurons remain poorly understood, growing evidence suggest a defective energetic metabolism in skeletal muscles participating in ALS-induced motor neuron death ultimately destabilizing neuromuscular junctions. In the present study, we report that a specific exercise paradigm, based on a high intensity and amplitude swimming exercise, significantly improves glucose metabolism in ALS mice. Using physiological tests and a biophysics approach based on nuclear magnetic resonance (NMR), we unexpectedly found that SOD1(G93A) ALS mice suffered from severe glucose intolerance, which was counteracted by high intensity swimming but not moderate intensity running exercise. Furthermore, swimming exercise restored the highly ALS-sensitive tibialis muscle through an autophagy-linked mechanism involving the expression of key glucose transporters and metabolic enzymes, including GLUT4 and glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Importantly, GLUT4 and GAPDH expression defects were also found in muscles from ALS patients. Moreover, we report that swimming exercise induced a triglyceride accumulation in ALS tibialis, likely resulting from an increase in the expression levels of lipid transporters and biosynthesis enzymes, notably DGAT1 and related proteins. All these data provide the first molecular basis for the differential effects of specific exercise type and intensity in ALS, calling for the use of physical exercise as an appropriate intervention to alleviate symptoms in this debilitating disease.
Collapse
Affiliation(s)
- Céline Desseille
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France
| | - Séverine Deforges
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France
| | - Olivier Biondi
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France
| | - Léo Houdebine
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France
| | - Domenico D'amico
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France
| | - Antonin Lamazière
- Laboratoire de lipidomique, Faculté de Médecine Pierre et Marie Curie - Hôpital Saint-Antoine, Université Paris 6, Paris, France
| | - Cédric Caradeuc
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,UMR 8601 CNRS, Université Paris Descartes, Paris, France
| | - Gildas Bertho
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,UMR 8601 CNRS, Université Paris Descartes, Paris, France
| | - Gaëlle Bruneteau
- Laboratoire de lipidomique, Faculté de Médecine Pierre et Marie Curie - Hôpital Saint-Antoine, Université Paris 6, Paris, France.,UMR 8601 CNRS, Université Paris Descartes, Paris, France
| | - Laure Weill
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France
| | - Jean Bastin
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France
| | - Fatima Djouadi
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France
| | - François Salachas
- Laboratoire de lipidomique, Faculté de Médecine Pierre et Marie Curie - Hôpital Saint-Antoine, Université Paris 6, Paris, France.,Hôpital de la Salpêtrière, Département des Maladies du Système Nerveux, Equipe Neurogénétique et Physiologie, Institut du Cerveau et de la Moelle, Paris, France
| | - Philippe Lopes
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France.,UFR Sciences Fondamentales Appliquées, Département STAPS, Université d'Evry-Val-d'Essonne, Evry, France
| | - Christophe Chanoine
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France
| | - Charbel Massaad
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France
| | - Frédéric Charbonnier
- Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Université Paris Descartes, Paris, France.,INSERM, UMR-S 1124, Paris, France
| |
Collapse
|
41
|
Zhang Y, Zhang Y, Ding GL, Liu XM, Ye J, Sheng JZ, Fan J, Huang HF. Regulation of hepatic pyruvate dehydrogenase phosphorylation in offspring glucose intolerance induced by intrauterine hyperglycemia. Oncotarget 2017; 8:15205-15212. [PMID: 28148899 PMCID: PMC5362479 DOI: 10.18632/oncotarget.14837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/11/2017] [Indexed: 01/07/2023] Open
Abstract
Aim Gestational diabetes mellitus (GDM) has been shown to be associated with a high risk of diabetes in offspring. In mitochondria, the inhibition of pyruvate dehydrogenase (PDH) activity by PDH phosphorylation is involved in the development of diabetes. We aimed to determine the role of PDH phosphorylation in the liver in GDM-induced offspring glucose intolerance. Results PDH phosphorylation was increased in lymphocytes from the umbilical cord blood of the GDM patients and in high glucose-treated hepatic cells. Both the male and female offspring from GDM mice had elevated liver weights and glucose intolerance. Further, PDH phosphorylation was increased in the livers of both the male and female offspring from GDM mice, and elevated acetylation may have contributed to this increased phosphorylation. Materials and methods We obtained lymphocytes from umbilical cord blood collected from both normal and GDM pregnant women. In addition, we obtained the offspring of streptozotocin-induced GDM female pregnant mice. The glucose tolerance test was performed to assess glucose tolerance in the offspring. Further, Western blotting was conducted to detect changes in protein levels. Conclusions Intrauterine hyperglycemia induced offspring glucose intolerance by inhibiting PDH activity, along with increased PDH phosphorylation in the liver, and this effect might be mediated by enhanced mitochondrial protein acetylation.
Collapse
Affiliation(s)
- Yong Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.,Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ying Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Lian Ding
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xin-Mei Liu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jianping Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Jian-Zhong Sheng
- Department of Pathophysiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jianxia Fan
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - He-Feng Huang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.,Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
42
|
Gudiksen A, Bertholdt L, Stankiewicz T, Tybirk J, Plomgaard P, Bangsbo J, Pilegaard H. Effects of training status on PDH regulation in human skeletal muscle during exercise. Pflugers Arch 2017; 469:1615-1630. [PMID: 28801776 DOI: 10.1007/s00424-017-2019-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/13/2017] [Accepted: 06/14/2017] [Indexed: 12/23/2022]
Abstract
Pyruvate dehydrogenase (PDH) is the gateway enzyme for carbohydrate-derived pyruvate feeding into the TCA cycle. PDH may play a central role in regulating substrate shifts during exercise, but the influence of training state on PDH regulation during exercise is not fully elucidated. The purpose of this study was to investigate the impact of training state on post-translational regulation of PDHa activity during submaximal and exhaustive exercise. Eight untrained and nine endurance exercise-trained healthy male subjects performed incremental exercise on a cycle ergometer: 40 min at 50% incremental peak power output (IPPO), 10 min at 65% (IPPO), followed by 80% (IPPO) until exhaustion. Trained subjects had higher (P < 0.05) PDH-E1α, PDK1, PDK2, PDK4, and PDP1 protein content as well as PDH phosphorylation and PDH acetylation. Exercising at the same relative intensity led to similar muscle PDH activation in untrained and trained subjects, whereas PDHa activity at exhaustion was higher (P < 0.05) in trained than untrained. Furthermore, exercise induced similar PDH dephosphorylation in untrained and trained subjects, while PDH acetylation was increased (P < 0.05) only in trained subjects. In conclusion, PDHa activity and PDH dephosphorylation were well adjusted to the relative exercise intensity during submaximal exercise. In addition, higher PDHa activity in trained than untrained at exhaustion seemed related to differences in glycogen utilization rather than differences in PDH phosphorylation and acetylation state, although site-specific contributions cannot be ruled out.
Collapse
Affiliation(s)
- Anders Gudiksen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, August Krogh Building, Universitetsparken 13, 2100, Copenhagen, Denmark
| | - Lærke Bertholdt
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, August Krogh Building, Universitetsparken 13, 2100, Copenhagen, Denmark
| | - Tomasz Stankiewicz
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, August Krogh Building, Universitetsparken 13, 2100, Copenhagen, Denmark
| | - Jonas Tybirk
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Peter Plomgaard
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bangsbo
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Pilegaard
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, August Krogh Building, Universitetsparken 13, 2100, Copenhagen, Denmark.
| |
Collapse
|
43
|
Jensen SR, Schoof EM, Wheeler SE, Hvid H, Ahnfelt-Rønne J, Hansen BF, Nishimura E, Olsen GS, Kislinger T, Brubaker PL. Quantitative Proteomics of Intestinal Mucosa From Male Mice Lacking Intestinal Epithelial Insulin Receptors. Endocrinology 2017; 158:2470-2485. [PMID: 28591806 DOI: 10.1210/en.2017-00194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/31/2017] [Indexed: 12/16/2022]
Abstract
The goal of the present study was to determine whether loss of the insulin receptor alters the molecular landscape of the intestinal mucosa, using intestinal-epithelial insulin receptor knockout (IE-irKO) mice and both genetic (IRfl/fl and Villin-cre) controls. Quantitative proteomic analysis by liquid chromatography mass spectrometry was applied to jejunal and colonic mucosa from mice fed a normal chow diet and mice fed a Western diet (WD). Jejunal mucosa from IE-irKO mice demonstrated alterations in all intestinal cell lineages: Paneth, goblet, absorptive, and enteroendocrine cells. Only goblet and absorptive cells were affected in the colon. Also, a marked effect of WD consumption was found on the gut proteome. A substantial reduction was detected in Paneth cell proteins with antimicrobial activity, including lysozyme C-1, angiogenin-4, cryptdin-related sequence 1C-3 and -2, α-defensin 17, and intelectin-1a. The key protein expressed by goblet cells, mucin-2, was also reduced in the IE-irKO mice. Proteins involved in lipid metabolism, including aldose reductase-related protein 1, 15-hydroxyprostaglandin dehydrogenase, apolipoprotein A-II, and pyruvate dehydrogenase kinase isozyme 4, were increased in the mucosa of WD-fed IE-irKO mice compared with controls. In contrast, expression of the nutrient-responsive gut hormones, glucose-dependent insulinotropic polypeptide and neurotensin, was reduced in the jejunal mucosa of IE-irKO mice, and the expression of proteins of the P-type adenosine triphosphatases and the solute carrier-transporter family was reduced in the colon of WD-fed IE-irKO mice. In conclusion, IE-irKO mice display a distinct molecular phenotype, suggesting a biological role of insulin and its receptor in determining differentiated cell specificity in the intestinal epithelium.
Collapse
Affiliation(s)
- Stina Rikke Jensen
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Metabolic Disease Research, Novo Nordisk A/S, Måløv DK-2760, Denmark
| | - Erwin M Schoof
- Princess Margaret Hospital Cancer Centre, University Health Network, Ontario M5G 2M9, Canada
| | - Sarah E Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Henning Hvid
- Metabolic Disease Research, Novo Nordisk A/S, Måløv DK-2760, Denmark
| | | | - Bo Falck Hansen
- Metabolic Disease Research, Novo Nordisk A/S, Måløv DK-2760, Denmark
| | - Erica Nishimura
- Metabolic Disease Research, Novo Nordisk A/S, Måløv DK-2760, Denmark
| | | | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Patricia L Brubaker
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
44
|
Tricò D, Prinsen H, Giannini C, de Graaf R, Juchem C, Li F, Caprio S, Santoro N, Herzog RI. Elevated α-Hydroxybutyrate and Branched-Chain Amino Acid Levels Predict Deterioration of Glycemic Control in Adolescents. J Clin Endocrinol Metab 2017; 102:2473-2481. [PMID: 28482070 PMCID: PMC5505187 DOI: 10.1210/jc.2017-00475] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/14/2017] [Indexed: 12/14/2022]
Abstract
CONTEXT Traditional risk factors for type 2 diabetes mellitus are weak predictors of changes in glucose tolerance and insulin sensitivity in youth. OBJECTIVE To identify early metabolic features of insulin resistance (IR) in youth and whether they predict deterioration of glycemic control. DESIGN AND SETTING A cross-sectional and longitudinal study was conducted at the Yale Pediatric Obesity Clinic. PATIENTS AND INTERVENTION Concentrations of α-hydroxybutyrate, β-hydroxybutyrate, lactate, and branched-chain amino acids (BCAAs) were measured by nuclear magnetic resonance spectroscopy in 78 nondiabetic adolescents during an oral glucose tolerance test (OGTT). Associations between baseline metabolic alterations and longitudinal changes in glucose control were tested in 16 subjects after a mean follow-up of 2.3 years. MAIN OUTCOME MEASURES The relationship between metabolite levels, parameters of IR, and glycemic control, and their progression over time. RESULTS Elevated fasting α-hydroxybutyrate levels were observed in adolescents with reduced insulin sensitivity after adjusting for age, sex, ethnicity, Tanner stage, and body mass index z-score (P = 0.014). Plasma α-hydroxybutyrate and BCAAs were increased throughout the course of the OGTT in this group (P < 0.03). Notably, borderline IR was associated with a progressive α-hydroxybutyrate decrease from elevated baseline concentrations to normal levels (P = 0.02). Increased baseline α-hydroxybutyrate concentrations were further associated with progressive worsening of glucose tolerance and disposition index. CONCLUSION α-Hydroxybutyrate and BCAA concentrations during an OGTT characterize insulin-resistant youth and predict worsening of glycemic control. These findings provide potential biomarkers for risk assessment of type 2 diabetes and new insights into IR pathogenesis.
Collapse
Affiliation(s)
- Domenico Tricò
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06519
| | - Hetty Prinsen
- Department of Diagnostic Radiology and Biomedical Imaging, Magnetic Resonance Research Center, Yale University School of Medicine, New Haven, Connecticut 06519
| | - Cosimo Giannini
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut 06519
| | - Robin de Graaf
- Department of Diagnostic Radiology and Biomedical Imaging, Magnetic Resonance Research Center, Yale University School of Medicine, New Haven, Connecticut 06519
| | - Christoph Juchem
- Department of Diagnostic Radiology and Biomedical Imaging, Magnetic Resonance Research Center, Yale University School of Medicine, New Haven, Connecticut 06519
| | - Fangyong Li
- Yale Center for Analytical Sciences, Yale University School of Public Health, New Haven, Connecticut 06519
| | - Sonia Caprio
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut 06519
| | - Nicola Santoro
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut 06519
- Dipartimento di Medicina e Scienze della Salute, Università degli Studi del Molise, Campobasso 86100, Italy
| | - Raimund I. Herzog
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06519
| |
Collapse
|
45
|
Tyszka-Czochara M, Bukowska-Strakova K, Majka M. Metformin and caffeic acid regulate metabolic reprogramming in human cervical carcinoma SiHa/HTB-35 cells and augment anticancer activity of Cisplatin via cell cycle regulation. Food Chem Toxicol 2017; 106:260-272. [PMID: 28576465 DOI: 10.1016/j.fct.2017.05.065] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/25/2017] [Accepted: 05/28/2017] [Indexed: 12/25/2022]
Abstract
Metformin shows benefits in anticancer prevention in humans. In this study, normal human fibroblasts (FB) and metastatic cervical cancer cells (SiHa) were exposed to 10 mM Metformin (Met), 100 μM Caffeic Acid (trans-3,4-dihydroxycinnamic acid, CA) or combination of the compounds. Both drugs were selectively toxic towards cancer cells, but neither Met nor CA treatment suppressed growth of normal cells. Met and CA regulated metabolic reprogramming in SiHa tumor cells through different mechanisms: Met suppressed regulatory enzymes Glurtaminase (GLS) and Malic Enzyme 1 (ME1) and enhanced pyruvate oxidation via tricarboxylic acids (TCA) cycle, while CA acted as glycolytic inhibitor. Met/CA treatment impaired expression of Sterol Regulatory Element-Binding Protein 1 (SREBP1c) which resulted in alleviation of de novo synthesis of unsaturated fatty acid. The toxic action of CisPt was supported by Met and CA not only in tumor cells, but also during co-culture of SiHa GFP+ cells with fibroblasts. Furthermore, Met and CA augmented Cisplatin (CisPt) action against quiescent tumor cells involving reprogramming of cell cycle. Our findings provide new insights into specific targeting of mitochondrial metabolism in neoplastic cells and into designing new cisplatin-based selective strategies for treating cervical cancer in humans with regard to the role of tumor microenvironment.
Collapse
Affiliation(s)
- Malgorzata Tyszka-Czochara
- Department of Radioligands, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland.
| | - Karolina Bukowska-Strakova
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka 265, 30-663 Krakow, Poland.
| | - Marcin Majka
- Department of Transplantation, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 258, 30-688 Krakow, Poland.
| |
Collapse
|
46
|
Han L, Shen WJ, Bittner S, Kraemer FB, Azhar S. PPARs: regulators of metabolism and as therapeutic targets in cardiovascular disease. Part I: PPAR-α. Future Cardiol 2017; 13:259-278. [PMID: 28581332 PMCID: PMC5941715 DOI: 10.2217/fca-2016-0059] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 03/21/2017] [Indexed: 02/07/2023] Open
Abstract
This article provides a comprehensive review about the molecular and metabolic actions of PPAR-α. It describes its structural features, ligand specificity, gene transcription mechanisms, functional characteristics and target genes. In addition, recent progress with the use of loss of function and gain of function mouse models in the discovery of diverse biological functions of PPAR-α, particularly in the vascular system and the status of the development of new single, dual, pan and partial PPAR agonists (PPAR modulators) in the clinical management of metabolic diseases are presented. This review also summarizes the clinical outcomes from a large number of clinical trials aimed at evaluating the atheroprotective actions of current clinically used PPAR-α agonists, fibrates and statin-fibrate combination therapy.
Collapse
Affiliation(s)
- Lu Han
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Wen-Jun Shen
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Stefanie Bittner
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Fredric B Kraemer
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Salman Azhar
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
47
|
Gudiksen A, Bertholdt L, Vingborg MB, Hansen HW, Ringholm S, Pilegaard H. Muscle interleukin-6 and fasting-induced PDH regulation in mouse skeletal muscle. Am J Physiol Endocrinol Metab 2017; 312:E204-E214. [PMID: 28028037 DOI: 10.1152/ajpendo.00291.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 11/22/2022]
Abstract
Fasting prompts a metabolic shift in substrate utilization from carbohydrate to predominant fat oxidation in skeletal muscle, and pyruvate dehydrogenase (PDH) is seen as a controlling link between the competitive oxidation of carbohydrate and fat during metabolic challenges like fasting. Interleukin (IL)-6 has been proposed to be released from muscle with concomitant effects on both glucose and fat utilization. The aim was to test the hypothesis that muscle IL-6 has a regulatory impact on fasting-induced suppression of skeletal muscle PDH. Skeletal muscle-specific IL-6 knockout (IL-6 MKO) mice and floxed littermate controls (control) were either fed or fasted for 6 or 18 h. Lack of muscle IL-6 elevated the respiratory exchange ratio in the fed and early fasting state, but not with prolonged fasting. Activity of PDH in the active form (PDHa) was higher in fed and fasted IL-6 MKO than in control mice at 18 h, but not at 6 h, whereas lack of muscle IL-6 did not prevent downregulation of PDHa activity in skeletal muscle or changes in plasma and muscle substrate levels in response to 18 h of fasting. Phosphorylation of three of four sites on PDH-E1α increased with 18 h of fasting, but was lower in IL-6 MKO mice than in control. In addition, both PDK4 mRNA and protein increased with 6 and 18 h of fasting in both genotypes, but PDK4 protein was lower in IL-6 MKO than in control. In conclusion, skeletal muscle IL-6 seems to regulate whole body substrate utilization in the fed, but not fasted, state and influence skeletal muscle PDHa activity in a circadian manner. However, skeletal muscle IL-6 is not required for maintaining metabolic flexibility in response to fasting.
Collapse
Affiliation(s)
- Anders Gudiksen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Laerke Bertholdt
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Birkkjaer Vingborg
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Watson Hansen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Stine Ringholm
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Pilegaard
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
Global view of cognate kinase activation by the human pyruvate dehydrogenase complex. Sci Rep 2017; 7:42760. [PMID: 28230160 PMCID: PMC5322387 DOI: 10.1038/srep42760] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/13/2017] [Indexed: 12/11/2022] Open
Abstract
The human pyruvate dehydrogenase complex (PDC) comprises four multidomain components, E1, E3, E2 and an E3-binding protein (E3BP), the latter two forming the core as E2·E3BP sub-complex. Pyruvate flux through PDC is regulated via phosphorylation (inactivation) at E1 by four PDC kinases (PDKs), and reactivation by two PDC phosphatases. Up-regulation of PDK isoform gene expression is reported in several forms of cancer, while PDKs may be further activated by PDC by binding to the E2·E3BP core. Hence, the PDK: E2·E3BP interaction provides new therapeutic targets. We carried out both functional kinetic and thermodynamic studies to demonstrate significant differences in the activation of PDK isoforms by binding to the E2·E3BP core: (i) PDK2 needs no activation by E2·E3BP for efficient functioning, while PDK4 was the least effective of the four isoforms, and could not be activated by E2·E3BP. Hence, development of inhibitors to the interaction of PDK2 and PDK4 with E2·E3BP is not promising; (ii) Design of inhibitors to interfere with interaction of E2·E3BP with PDK1 and PDK3 is promising. PDK3 needs E2·E3BP core for activation, an activation best achieved by synergistic combination of E2-derived catalytic domain and tridomain.
Collapse
|
49
|
Caffeic Acid Expands Anti-Tumor Effect of Metformin in Human Metastatic Cervical Carcinoma HTB-34 Cells: Implications of AMPK Activation and Impairment of Fatty Acids De Novo Biosynthesis. Int J Mol Sci 2017; 18:ijms18020462. [PMID: 28230778 PMCID: PMC5343995 DOI: 10.3390/ijms18020462] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 02/07/2023] Open
Abstract
The efficacy of cancer treatments is often limited and associated with substantial toxicity. Appropriate combination of drug targeting specific mechanisms may regulate metabolism of tumor cells to reduce cancer cell growth and to improve survival. Therefore, we investigated the effects of anti-diabetic drug Metformin (Met) and a natural compound caffeic acid (trans-3,4-dihydroxycinnamic acid, CA) alone and in combination to treat an aggressive metastatic human cervical HTB-34 (ATCC CRL1550) cancer cell line. CA at concentration of 100 µM, unlike Met at 10 mM, activated 5'-adenosine monophosphate-activated protein kinase (AMPK). What is more, CA contributed to the fueling of mitochondrial tricarboxylic acids (TCA) cycle with pyruvate by increasing Pyruvate Dehydrogenase Complex (PDH) activity, while Met promoted glucose catabolism to lactate. Met downregulated expression of enzymes of fatty acid de novo synthesis, such as ATP Citrate Lyase (ACLY), Fatty Acid Synthase (FAS), Fatty Acyl-CoA Elongase 6 (ELOVL6), and Stearoyl-CoA Desaturase-1 (SCD1) in cancer cells. In conclusion, CA mediated reprogramming of glucose processing through TCA cycle via oxidative decarboxylation. The increased oxidative stress, as a result of CA treatment, sensitized cancer cells and, acting on cell biosynthesis and bioenergetics, made HTB-34 cells more susceptible to Met and successfully inhibited neoplastic cells. The combination of Metformin and caffeic acid to suppress cervical carcinoma cells by two independent mechanisms may provide a promising approach to cancer treatment.
Collapse
|
50
|
Brough PA, Baker L, Bedford S, Brown K, Chavda S, Chell V, D'Alessandro J, Davies NGM, Davis B, Le Strat L, Macias AT, Maddox D, Mahon PC, Massey AJ, Matassova N, McKenna S, Meissner JWG, Moore JD, Murray JB, Northfield CJ, Parry C, Parsons R, Roughley SD, Shaw T, Simmonite H, Stokes S, Surgenor A, Stefaniak E, Robertson A, Wang Y, Webb P, Whitehead N, Wood M. Application of Off-Rate Screening in the Identification of Novel Pan-Isoform Inhibitors of Pyruvate Dehydrogenase Kinase. J Med Chem 2017; 60:2271-2286. [PMID: 28199108 DOI: 10.1021/acs.jmedchem.6b01478] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Libraries of nonpurified resorcinol amide derivatives were screened by surface plasmon resonance (SPR) to determine the binding dissociation constant (off-rate, kd) for compounds binding to the pyruvate dehydrogenase kinase (PDHK) enzyme. Parallel off-rate measurements against HSP90 and application of structure-based drug design enabled rapid hit to lead progression in a program to identify pan-isoform ATP-competitive inhibitors of PDHK. Lead optimization identified selective sub-100-nM inhibitors of the enzyme which significantly reduced phosphorylation of the E1α subunit in the PC3 cancer cell line in vitro.
Collapse
Affiliation(s)
- Paul A Brough
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Lisa Baker
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Simon Bedford
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Kirsten Brown
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Seema Chavda
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Victoria Chell
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | | | | | - Ben Davis
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Loic Le Strat
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Alba T Macias
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Daniel Maddox
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Patrick C Mahon
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Andrew J Massey
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Natalia Matassova
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Sean McKenna
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | | | - Jonathan D Moore
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - James B Murray
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | | | - Charles Parry
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Rachel Parsons
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Stephen D Roughley
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Terry Shaw
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Heather Simmonite
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Stephen Stokes
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Allan Surgenor
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Emma Stefaniak
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Alan Robertson
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Yikang Wang
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Paul Webb
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Neil Whitehead
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| | - Mike Wood
- Vernalis (R&D) Ltd. , Granta Park, Great Abington, Cambridge CB21 6GB, U.K
| |
Collapse
|