101
|
Ng SP, Nomura W, Takahashi H, Inoue K, Kawada T, Goto T. Methylglyoxal attenuates isoproterenol-induced increase in uncoupling protein 1 expression through activation of JNK signaling pathway in beige adipocytes. Biochem Biophys Rep 2021; 28:101127. [PMID: 34527816 PMCID: PMC8430270 DOI: 10.1016/j.bbrep.2021.101127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/27/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Methylglyoxal (MG) is a metabolite derived from glycolysis whose levels in the blood and tissues of patients with diabetes are higher than those of healthy individuals, suggesting that MG is associated with the development of diabetic complications. However, it remains unknown whether high levels of MG are a cause or consequence of diabetes. Here, we show that MG negatively affects the expression of uncoupling protein 1 (UCP1), which is involved in thermogenesis and the regulation of systemic metabolism. Decreased Ucp1 expression is associated with obesity and type 2 diabetes. We found that MG attenuated the increase in Ucp1 expression following treatment with isoproterenol in beige adipocytes. However, MG did not affect protein kinase A signaling, the core coordinator of isoproterenol-induced Ucp1 expression. Instead, MG activated c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinases. We found that JNK inhibition, but not p38, recovered isoproterenol-stimulated Ucp1 expression under MG treatment. Altogether, these results suggest an inhibitory role of MG on the thermogenic function of beige adipocytes through the JNK signaling pathway.
Collapse
Key Words
- BBGC, S-p-bromobenzylglutathione cyclopentyl diester
- Beige adipocytes
- CREB, cAMP response element-binding protein
- ERK, extracellular receptor kinase
- HSL, hormone-sensitive lipase
- JNK
- JNK, c-Jun N-terminal kinase
- MG, methylglyoxal
- Methylglyoxal
- NAC, N-acetyl-l-cysteine
- NEFA, non-esterified fatty acids
- PKA, protein kinase A
- SEM, standard error of the mean
- Ucp1
- iWAT, inguinal white adipose tissue
Collapse
Affiliation(s)
- Su-Ping Ng
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Wataru Nomura
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8317, Japan
- Corresponding author. Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan.
| | - Haruya Takahashi
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Kazuo Inoue
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8317, Japan
| | - Teruo Kawada
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8317, Japan
| | - Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8317, Japan
- Corresponding author. Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan.
| |
Collapse
|
102
|
Yang Y, Zhang H, Guo Z, Zou S, Long F, Wu J, Li P, Zhao GP, Zhao W. Global Insights Into Lysine Acylomes Reveal Crosstalk Between Lysine Acetylation and Succinylation in Streptomyces coelicolor Metabolic Pathways. Mol Cell Proteomics 2021; 20:100148. [PMID: 34530157 PMCID: PMC8498004 DOI: 10.1016/j.mcpro.2021.100148] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/08/2021] [Indexed: 02/09/2023] Open
Abstract
Lysine acylations are reversible and ubiquitous post-translational modifications that play critical roles in regulating multiple cellular processes. In the current study, highly abundant and dynamic acetylation, besides succinylation, was uncovered in a soil bacterium, Streptomyces coelicolor. By affinity enrichment using anti–acetyl-lysine antibody and the following LC−MS/MS analysis, a total of 1298 acetylation sites among 601 proteins were identified. Bioinformatics analyses suggested that these acetylated proteins have diverse subcellular localization and were enriched in a wide range of biological functions. Specifically, a majority of the acetylated proteins were also succinylated in the tricarboxylic acid cycle and protein translation pathways, and the bimodification occurred at the same sites in some proteins. The acetylation and succinylation sites were quantified by knocking out either the deacetylase ScCobB1 or the desuccinylase ScCobB2, demonstrating a possible competitive relationship between the two acylations. Moreover, in vitro experiments using synthetically modified peptides confirmed the regulatory crosstalk between the two sirtuins, which may be involved in the collaborative regulation of cell physiology. Collectively, these results provided global insights into the S. coelicolor acylomes and laid a foundation for characterizing the regulatory roles of the crosstalk between lysine acetylation and succinylation in the future. A highly abundant and dynamic acetylation is discovered in Streptomyces coelicolor. Quantitative acetylome and succinylome analyses in Streptomyces coelicolor. The bimodification proteins are enriched in multiple metabolic pathways.
Collapse
Affiliation(s)
- Yujiao Yang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, China; CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Key Laboratory of Synthetic Biology, University of Chinese Academy of Sciences, Beijing, China
| | - Hong Zhang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhenyang Guo
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Siwei Zou
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fei Long
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jiacheng Wu
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, China; Key Laboratory of Synthetic Biology, University of Chinese Academy of Sciences, Beijing, China
| | - Peng Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
| | - Guo-Ping Zhao
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, China; CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Key Laboratory of Synthetic Biology, University of Chinese Academy of Sciences, Beijing, China.
| | - Wei Zhao
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
103
|
Fernández-Veledo S, Ceperuelo-Mallafré V, Vendrell J. Rethinking succinate: an unexpected hormone-like metabolite in energy homeostasis. Trends Endocrinol Metab 2021; 32:680-692. [PMID: 34301438 DOI: 10.1016/j.tem.2021.06.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023]
Abstract
There has been an explosion of interest in the signaling capacity of energy metabolites. A prime example is the Krebs cycle substrate succinate, an archetypal respiratory substrate with functions beyond energy production as an intracellular and extracellular signaling molecule. Long associated with inflammation, emerging evidence supports a key role for succinate in metabolic processes relating to energy management. As the natural ligand for SUCNR1, a G protein-coupled receptor, succinate is akin to hormones and likely functions as a reporter of metabolism and stress. In this review, we reconcile new and old observations to outline a regulatory role for succinate in metabolic homeostasis. We highlight the importance of the succinate-SUCNR1 axis in metabolic diseases as an integrator of macrophage immune response, and we discuss new metabolic functions recently ascribed to succinate in specific tissues. Because circulating succinate has emerged as a promising biomarker in chronic metabolic diseases, a better understanding of the physiopathological role of the succinate-SUCNR1 axis in metabolism might open new avenues for clinical use in patients with obesity or diabetes.
Collapse
Affiliation(s)
- Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.
| | - Victòria Ceperuelo-Mallafré
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine and Surgery, University Rovira I Virgili, Tarragona, Spain
| | - Joan Vendrell
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine and Surgery, University Rovira I Virgili, Tarragona, Spain
| |
Collapse
|
104
|
Liu J, Shangguan Y, Tang D, Dai Y. Histone succinylation and its function on the nucleosome. J Cell Mol Med 2021; 25:7101-7109. [PMID: 34160884 PMCID: PMC8335665 DOI: 10.1111/jcmm.16676] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/01/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023] Open
Abstract
Protein post‐translational modifications (PTMs) of histones are ubiquitous regulatory mechanisms involved in many biological processes, including replication, transcription, DNA damage repair and ontogenesis. Recently, many short‐chain acylation histone modifications have been identified by mass spectrometry (MS). Lysine succinylation (Ksuc or Ksucc) is a newly identified histone PTM that changes the chemical environment of histones and is similar to other acylation modifications; lysine succinylation appears to accumulate at transcriptional start sites and to correlate with gene expression. Although numerous studies are ongoing, there is a lack of reviews on the Ksuc of histones. Here, we review lysine succinylation sites on histones, including the chemical characteristics and the mechanism by which lysine succinylation influences nucleosomal structure, chromatin dynamics and several diseases and then discuss lysine succinylation regulation to identify theoretical and experimental proof of Ksuc on histones and in diseases to inspire further research into histone lysine succinylation as a target of disease treatment in the future.
Collapse
Affiliation(s)
- Jiayi Liu
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital(Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.,School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yu Shangguan
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital(Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.,Guangxi Key Laboratory of Metabolic Disease Research, Central Laboratory of Guilin, 924st Hospital, Guilin, China
| | - Donge Tang
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital(Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.,School of Medicine, Southern University of Science and Technology, Shenzhen, China.,Guangxi Key Laboratory of Metabolic Disease Research, Central Laboratory of Guilin, 924st Hospital, Guilin, China
| | - Yong Dai
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital(Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.,School of Medicine, Southern University of Science and Technology, Shenzhen, China.,Guangxi Key Laboratory of Metabolic Disease Research, Central Laboratory of Guilin, 924st Hospital, Guilin, China
| |
Collapse
|
105
|
Cohen P, Kajimura S. The cellular and functional complexity of thermogenic fat. Nat Rev Mol Cell Biol 2021; 22:393-409. [PMID: 33758402 PMCID: PMC8159882 DOI: 10.1038/s41580-021-00350-0] [Citation(s) in RCA: 279] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2021] [Indexed: 02/01/2023]
Abstract
Brown and beige adipocytes are mitochondria-enriched cells capable of dissipating energy in the form of heat. These thermogenic fat cells were originally considered to function solely in heat generation through the action of the mitochondrial protein uncoupling protein 1 (UCP1). In recent years, significant advances have been made in our understanding of the ontogeny, bioenergetics and physiological functions of thermogenic fat. Distinct subtypes of thermogenic adipocytes have been identified with unique developmental origins, which have been increasingly dissected in cellular and molecular detail. Moreover, several UCP1-independent thermogenic mechanisms have been described, expanding the role of these cells in energy homeostasis. Recent studies have also delineated roles for these cells beyond the regulation of thermogenesis, including as dynamic secretory cells and as a metabolic sink. This Review presents our current understanding of thermogenic adipocytes with an emphasis on their development, biological functions and roles in systemic physiology.
Collapse
Affiliation(s)
- Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA.
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Department of Cell and Tissue Biology, UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
106
|
Sun L, Zhang H, Gao P. Metabolic reprogramming and epigenetic modifications on the path to cancer. Protein Cell 2021; 13:877-919. [PMID: 34050894 PMCID: PMC9243210 DOI: 10.1007/s13238-021-00846-7] [Citation(s) in RCA: 341] [Impact Index Per Article: 85.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Metabolic rewiring and epigenetic remodeling, which are closely linked and reciprocally regulate each other, are among the well-known cancer hallmarks. Recent evidence suggests that many metabolites serve as substrates or cofactors of chromatin-modifying enzymes as a consequence of the translocation or spatial regionalization of enzymes or metabolites. Various metabolic alterations and epigenetic modifications also reportedly drive immune escape or impede immunosurveillance within certain contexts, playing important roles in tumor progression. In this review, we focus on how metabolic reprogramming of tumor cells and immune cells reshapes epigenetic alterations, in particular the acetylation and methylation of histone proteins and DNA. We also discuss other eminent metabolic modifications such as, succinylation, hydroxybutyrylation, and lactylation, and update the current advances in metabolism- and epigenetic modification-based therapeutic prospects in cancer.
Collapse
Affiliation(s)
- Linchong Sun
- Guangzhou First People's Hospital, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, 510006, China.
| | - Huafeng Zhang
- The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230027, China. .,CAS Centre for Excellence in Cell and Molecular Biology, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Ping Gao
- Guangzhou First People's Hospital, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, 510006, China. .,School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China. .,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China.
| |
Collapse
|
107
|
Wang Z, Wang QA, Liu Y, Jiang L. Energy metabolism in brown adipose tissue. FEBS J 2021; 288:3647-3662. [PMID: 34028971 DOI: 10.1111/febs.16015] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 12/14/2022]
Abstract
Brown adipose tissue (BAT) is well known to burn calories through uncoupled respiration, producing heat to maintain body temperature. This 'calorie wasting' feature makes BAT a special tissue, which can function as an 'energy sink' in mammals. While a combination of high energy intake and low energy expenditure is the leading cause of overweight and obesity in modern society, activating a safe 'energy sink' has been proposed as a promising obesity treatment strategy. Metabolically, lipids and glucose have been viewed as the major energy substrates in BAT, while succinate, lactate, branched-chain amino acids, and other metabolites can also serve as energy substrates for thermogenesis. Since the cataplerotic and anaplerotic reactions of these metabolites interconnect with each other, BAT relies on its dynamic, flexible, and complex metabolism to support its special function. In this review, we summarize how BAT orchestrates the metabolic utilization of various nutrients to support thermogenesis and contributes to whole-body metabolic homeostasis.
Collapse
Affiliation(s)
- Zhichao Wang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA, USA
| | - Qiong A Wang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA, USA.,Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Frontier Science Center for Immunology and Metabolism, Institute for Advanced Studies, Wuhan University, China
| | - Lei Jiang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA, USA.,Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| |
Collapse
|
108
|
Michurina SS, Stafeev IS, Menshikov MY, Parfyonova YV. Mitochondrial dynamics keep balance of nutrient combustion in thermogenic adipocytes. Mitochondrion 2021; 59:157-168. [PMID: 34010673 DOI: 10.1016/j.mito.2021.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/02/2021] [Accepted: 05/13/2021] [Indexed: 12/21/2022]
Abstract
Non-shivering thermogenesis takes place in brown and beige adipocytes and facilitates cold tolerance and acclimation. However, thermogenesis in adipose tissue also was found to be activated in metabolic overload states for fast utilization of nutrients excess. This observation spurred research interest in mechanisms of thermogenesis regulation for metabolic overload and obesity prevention. One of proposed regulators of thermogenic efficiency in adipocytes is the dynamics of mitochondria, where thermogenesis takes place. Indeed, brown and beige adipocytes exhibit fragmented round-shaped mitochondria, while white adipocytes have elongated organelles with high ATP synthesis. Mitochondrial morphology can determine uncoupling protein 1 (UCP1) content, efficiency of catabolic pathways and electron transport chain, supplying thermogenesis. This review will highlight the co-regulation of mitochondrial dynamics and thermogenesis and formulate hypothetical ways for excessive nutrients burning in response to mitochondrial morphology manipulation.
Collapse
Affiliation(s)
- S S Michurina
- Lomonosov Moscow State University, 119234 Moscow, Russia; Institute of Experimental Cardiology, National Medical Research Centre for Cardiology, 121500 Moscow, Russia.
| | - I S Stafeev
- Institute of Experimental Cardiology, National Medical Research Centre for Cardiology, 121500 Moscow, Russia.
| | - M Y Menshikov
- Institute of Experimental Cardiology, National Medical Research Centre for Cardiology, 121500 Moscow, Russia
| | - Ye V Parfyonova
- Lomonosov Moscow State University, 119234 Moscow, Russia; Institute of Experimental Cardiology, National Medical Research Centre for Cardiology, 121500 Moscow, Russia
| |
Collapse
|
109
|
Molinari F, Feraco A, Mirabilii S, Saladini S, Sansone L, Vernucci E, Tomaselli G, Marzolla V, Rotili D, Russo MA, Ricciardi MR, Tafuri A, Mai A, Caprio M, Tafani M, Armani A. SIRT5 Inhibition Induces Brown Fat-Like Phenotype in 3T3-L1 Preadipocytes. Cells 2021; 10:cells10051126. [PMID: 34066961 PMCID: PMC8148511 DOI: 10.3390/cells10051126] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022] Open
Abstract
Brown adipose tissue (BAT) activity plays a key role in regulating systemic energy. The activation of BAT results in increased energy expenditure, making this tissue an attractive pharmacological target for therapies against obesity and type 2 diabetes. Sirtuin 5 (SIRT5) affects BAT function by regulating adipogenic transcription factor expression and mitochondrial respiration. We analyzed the expression of SIRT5 in the different adipose depots of mice. We treated 3T3-L1 preadipocytes and mouse primary preadipocyte cultures with the SIRT5 inhibitor MC3482 and investigated the effects of this compound on adipose differentiation and function. The administration of MC3482 during the early stages of differentiation promoted the expression of brown adipocyte and mitochondrial biogenesis markers. Upon treatment with MC3482, 3T3-L1 adipocytes showed an increased activation of the AMP-activated protein kinase (AMPK), which is known to stimulate brown adipocyte differentiation. This effect was paralleled by an increase in autophagic/mitophagic flux and a reduction in lipid droplet size, mediated by a higher lipolytic rate. Of note, MC3482 increased the expression and the activity of adipose triglyceride lipase, without modulating hormone-sensitive lipase. Our findings reveal that SIRT5 inhibition stimulates brown adipogenesis in vitro, supporting this approach as a strategy to stimulate BAT and counteract obesity.
Collapse
Affiliation(s)
- Francesca Molinari
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (F.M.); (S.S.); (M.T.)
| | - Alessandra Feraco
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00163 Rome, Italy; (A.F.); (V.M.); (M.C.)
| | - Simone Mirabilii
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
| | - Serena Saladini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (F.M.); (S.S.); (M.T.)
| | - Luigi Sansone
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele, 00166 Rome, Italy; (L.S.); (G.T.); (M.A.R.)
| | - Enza Vernucci
- Department of Cardiovascular, Nephrologic, Anesthesiologic and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy;
| | - Giada Tomaselli
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele, 00166 Rome, Italy; (L.S.); (G.T.); (M.A.R.)
| | - Vincenzo Marzolla
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00163 Rome, Italy; (A.F.); (V.M.); (M.C.)
| | - Dante Rotili
- Department of Chemistry and Technology of Drugs, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy; (D.R.); (A.M.)
| | - Matteo A. Russo
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele, 00166 Rome, Italy; (L.S.); (G.T.); (M.A.R.)
- MEBIC Consortium, San Raffaele Rome Open University, 00166 Rome, Italy
| | - Maria Rosaria Ricciardi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
| | - Agostino Tafuri
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
- Hematology, “Sant’ Andrea” University Hospital, Sapienza University of Rome, 00189 Rome, Italy
| | - Antonello Mai
- Department of Chemistry and Technology of Drugs, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy; (D.R.); (A.M.)
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00163 Rome, Italy; (A.F.); (V.M.); (M.C.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (F.M.); (S.S.); (M.T.)
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00163 Rome, Italy; (A.F.); (V.M.); (M.C.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Correspondence:
| |
Collapse
|
110
|
Abstract
Intermittent fasting, which can effectively reduce obesity and improve the related metabolic syndrome has become an exciting research area in recent years. Adipose tissue is pivotal in regulating the metabolism and determining the occurrence of obesity. In the current study, we aimed to investigate the effects of acute fasting (AF) on fat tissue. Mice were subjected to AF for 36 h, receiving normal chow (low-fat diet [LFD]) or a high-fat diet (HFD), with free ad libitum access to drinking water, and those fed on free-diet counterparts without fasting serveding as controls. We found that AF obviously reshaped the morphology of fat tissue (WAT) and promoted the beiging of white adipose tissue in both LFD- and HFD-fed mice. AF principally improved the lipid metabolism, and increased the M2- polarization of macrophages in WAT white fat tissue of HFD-fed mice. Interestingly, we found that AF dramatically upregulated Sirt5 expression levels and fat tissue succinylation, suggesting that AF-induced beneficial effects on fat might occur via the regulation of Sirt5 levels and altered succinylation in fatty tissues. Our study clearly showed the remodeling function of adipose tissue during AF; in terms of mechanism, the regulation of succinylation levels by AF might provide new insights into the mechanism(s) underlying the improvement in fat metabolism by energy restriction.
Collapse
Affiliation(s)
- Tuohua Mao
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Quanwei Wei
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Fang Zhao
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Chuanhai Zhang
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| |
Collapse
|
111
|
Shinde AB, Song A, Wang QA. Brown Adipose Tissue Heterogeneity, Energy Metabolism, and Beyond. Front Endocrinol (Lausanne) 2021; 12:651763. [PMID: 33953697 PMCID: PMC8092391 DOI: 10.3389/fendo.2021.651763] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/18/2021] [Indexed: 01/19/2023] Open
Abstract
Brown adipocyte in brown adipose tissue (BAT) specializes in expending energy through non-shivering thermogenesis, a process that produces heat either by uncoupling protein 1 (UCP1) dependent uncoupling of mitochondrial respiration or by UCP1 independent mechanisms. Apart from this, there is ample evidence suggesting that BAT has an endocrine function. Studies in rodents point toward its vital roles in glucose and lipid homeostasis, making it an important therapeutic target for treating metabolic disorders related to morbidities such as obesity and type 2 diabetes. The rediscovery of thermogenically active BAT depots in humans by several independent research groups in the last decade has revitalized interest in BAT as an even more promising therapeutic intervention. Over the last few years, there has been overwhelming interest in understanding brown adipocyte's developmental lineages and how brown adipocyte uniquely utilizes energy beyond UCP1 mediated uncoupling respiration. These new discoveries would be leveraged for designing novel therapeutic interventions for metabolic disorders.
Collapse
Affiliation(s)
- Abhijit Babaji Shinde
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, United States
| | - Anying Song
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, United States
| | - Qiong A. Wang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, United States
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, United States
| |
Collapse
|
112
|
Wang M, Lin H. Understanding the Function of Mammalian Sirtuins and Protein Lysine Acylation. Annu Rev Biochem 2021; 90:245-285. [PMID: 33848425 DOI: 10.1146/annurev-biochem-082520-125411] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein lysine acetylation is an important posttranslational modification that regulates numerous biological processes. Targeting lysine acetylation regulatory factors, such as acetyltransferases, deacetylases, and acetyl-lysine recognition domains, has been shown to have potential for treating human diseases, including cancer and neurological diseases. Over the past decade, many other acyl-lysine modifications, such as succinylation, crotonylation, and long-chain fatty acylation, have also been investigated and shown to have interesting biological functions. Here, we provide an overview of the functions of different acyl-lysine modifications in mammals. We focus on lysine acetylation as it is well characterized, and principles learned from acetylation are useful for understanding the functions of other lysine acylations. We pay special attention to the sirtuins, given that the study of sirtuins has provided a great deal of information about the functions of lysine acylation. We emphasize the regulation of sirtuins to illustrate that their regulation enables cells to respond to various signals and stresses.
Collapse
Affiliation(s)
- Miao Wang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA;
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA; .,Howard Hughes Medical Institute, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
113
|
Nicholls DG. Mitochondrial proton leaks and uncoupling proteins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2021; 1862:148428. [PMID: 33798544 DOI: 10.1016/j.bbabio.2021.148428] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/15/2021] [Accepted: 03/25/2021] [Indexed: 01/02/2023]
Abstract
Non-shivering thermogenesis in brown adipose tissue is mediated by uncoupling protein 1 (UCP1), which provides a carefully regulated proton re-entry pathway across the mitochondrial inner membrane operating in parallel to the ATP synthase and allowing respiration, and hence thermogenesis, to be released from the constraints of respiratory control. In the 40 years since UCP1 was first described, an extensive, and frequently contradictory, literature has accumulated, focused on the acute physiological regulation of the protein by fatty acids, purine nucleotides and possible additional factors. The purpose of this review is to examine, in detail, the experimental evidence underlying these proposed mechanisms. Emphasis will be placed on the methodologies employed and their relation to the physiological constraints under which the protein functions in the intact cell. The nature of the endogenous, UCP1-independent, proton leak will also be discussed. Finally, the troubled history of the putative novel uncoupling proteins, UCP2 and UCP3, will be evaluated.
Collapse
|
114
|
Liu J, Tan Y, Ao H, Feng W, Peng C. Aqueous extracts of Aconite promote thermogenesis in rats with hypothermia via regulating gut microbiota and bile acid metabolism. Chin Med 2021; 16:29. [PMID: 33741035 PMCID: PMC7980327 DOI: 10.1186/s13020-021-00437-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 03/08/2021] [Indexed: 12/15/2022] Open
Abstract
Background Intermittent or prolonged exposure to severe cold stress disturbs energy homeostasis and can lead to hypothermia, heart failure, Alzheimer’s disease, and so on. As the typical “hot” traditional Chinese medicine, Aconite has been widely used to treat cold-associated diseases for thousands of years, but its critical mechanisms for the promotion of thermogenesis are not fully resolved. Gut microbiota and its metabolites play a crucial role in maintaining energy homeostasis. Here, we investigated whether the aqueous extracts of Aconite (AA) can enhance thermogenesis through modulation of the composition and metabolism of gut microbiota in hypothermic rats. Methods The therapeutic effects of AA on body temperature, energy intake, and the histopathology of white adipose tissue and brown adipose tissue of hypothermic rats were assessed. Microbiota analysis based on 16 S rRNA and targeted metabolomics for bile acids (BAs) were used to evaluate the composition of gut microbiota and BAs pool. The antibiotic cocktail treatment was adopted to further confirm the relationship between the gut microbiota and the thermogenesis-promoting effects of AA. Results Our results showed a sharp drop in rectal temperature and body surface temperature in hypothermic rats. Administration of AA can significantly increase core body temperature, surface body temperature, energy intake, browning of white adipose tissue, and thermogenesis of brown adipose tissue. Importantly, these ameliorative effects of AA were accompanied by the shift of the disturbed composition of gut microbiota toward a healthier profile and the increased levels of BAs. In addition, the depletion of gut microbiota and the reduction of BAs caused by antibiotic cocktails reduced the thermogenesis-promoting effect of AA. Conclusions Our results demonstrated that AA promoted thermogenesis in rats with hypothermia via regulating gut microbiota and BAs metabolism. Our findings can also provide a novel solution for the treatment of thermogenesis-associated diseases such as rheumatoid arthritis, obesity, and type 2 diabetes. ![]()
Collapse
Affiliation(s)
- Juan Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China.,National Key Laboratory Breeding Base of Systematic Research, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Yuzhu Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Hui Ao
- National Key Laboratory Breeding Base of Systematic Research, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Wuwen Feng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China. .,National Key Laboratory Breeding Base of Systematic Research, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China.
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China. .,National Key Laboratory Breeding Base of Systematic Research, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China.
| |
Collapse
|
115
|
Maissan P, Mooij EJ, Barberis M. Sirtuins-Mediated System-Level Regulation of Mammalian Tissues at the Interface between Metabolism and Cell Cycle: A Systematic Review. BIOLOGY 2021; 10:194. [PMID: 33806509 PMCID: PMC7999230 DOI: 10.3390/biology10030194] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
Sirtuins are a family of highly conserved NAD+-dependent proteins and this dependency links Sirtuins directly to metabolism. Sirtuins' activity has been shown to extend the lifespan of several organisms and mainly through the post-translational modification of their many target proteins, with deacetylation being the most common modification. The seven mammalian Sirtuins, SIRT1 through SIRT7, have been implicated in regulating physiological responses to metabolism and stress by acting as nutrient sensors, linking environmental and nutrient signals to mammalian metabolic homeostasis. Furthermore, mammalian Sirtuins have been implicated in playing major roles in mammalian pathophysiological conditions such as inflammation, obesity and cancer. Mammalian Sirtuins are expressed heterogeneously among different organs and tissues, and the same holds true for their substrates. Thus, the function of mammalian Sirtuins together with their substrates is expected to vary among tissues. Any therapy depending on Sirtuins could therefore have different local as well as systemic effects. Here, an introduction to processes relevant for the actions of Sirtuins, such as metabolism and cell cycle, will be followed by reasoning on the system-level function of Sirtuins and their substrates in different mammalian tissues. Their involvement in the healthy metabolism and metabolic disorders will be reviewed and critically discussed.
Collapse
Affiliation(s)
- Parcival Maissan
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
| | - Eva J. Mooij
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| |
Collapse
|
116
|
Gaspar RC, Pauli JR, Shulman GI, Muñoz VR. An update on brown adipose tissue biology: a discussion of recent findings. Am J Physiol Endocrinol Metab 2021; 320:E488-E495. [PMID: 33459179 PMCID: PMC7988785 DOI: 10.1152/ajpendo.00310.2020] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/20/2022]
Abstract
Brown adipose tissue (BAT) has been encouraged as a potential treatment for obesity and comorbidities due to its thermogenic activity capacity and contribution to energy expenditure. Some interventions such as cold and β-adrenergic drugs are able to activate BAT thermogenesis as well as promote differentiation of white adipocytes into brown-like cells (browning), enhancing the thermogenic activity of these cells. In this mini-review, we discuss new mechanisms related to BAT and energy expenditure. In this regard, we will also discuss recent studies that have revealed the existence of important secretory molecules from BAT "batokines" that act in autocrine, paracrine, and endocrine mechanisms, which in turn may explain some of the beneficial roles of BAT on whole body glucose and fat metabolism. Finally, we will discuss new insights related to BAT thermogenesis with an additional focus on the distinct features of BAT metabolism between rodents and humans.
Collapse
Affiliation(s)
- Rafael C Gaspar
- Department of Health Sciences, Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, São Paulo, Brazil
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - José R Pauli
- Department of Health Sciences, Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, São Paulo, Brazil
- OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Vitor R Muñoz
- Department of Health Sciences, Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, São Paulo, Brazil
| |
Collapse
|
117
|
Shi M, Huang XY, Ren XY, Wei XY, Ma Y, Lin ZZ, Liu DT, Song L, Zhao TJ, Li G, Yao L, Zhu M, Zhang C, Xie C, Wu Y, Wu HM, Fan LP, Ou J, Zhan YH, Lin SY, Lin SC. AIDA directly connects sympathetic innervation to adaptive thermogenesis by UCP1. Nat Cell Biol 2021; 23:268-277. [PMID: 33664495 DOI: 10.1038/s41556-021-00642-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023]
Abstract
The sympathetic nervous system-catecholamine-uncoupling protein 1 (UCP1) axis plays an essential role in non-shivering adaptive thermogenesis. However, whether there exists a direct effector that physically connects catecholamine signalling to UCP1 in response to acute cold is unknown. Here we report that outer mitochondrial membrane-located AIDA is phosphorylated at S161 by the catecholamine-activated protein kinase A (PKA). Phosphorylated AIDA translocates to the intermembrane space, where it binds to and activates the uncoupling activity of UCP1 by promoting cysteine oxidation of UCP1. Adipocyte-specific depletion of AIDA abrogates UCP1-dependent thermogenesis, resulting in hypothermia during acute cold exposure. Re-expression of S161A-AIDA, unlike wild-type AIDA, fails to restore the acute cold response in Aida-knockout mice. The PKA-AIDA-UCP1 axis is highly conserved in mammals, including hibernators. Denervation of the sympathetic postganglionic fibres abolishes cold-induced AIDA-dependent thermogenesis. These findings uncover a direct mechanistic link between sympathetic input and UCP1-mediated adaptive thermogenesis.
Collapse
Affiliation(s)
- Meng Shi
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xiao-Yu Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xin-Yi Ren
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xiao-Yan Wei
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yue Ma
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Zhi-Zhong Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Dong-Tai Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Lintao Song
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Tong-Jin Zhao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Guang Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Luming Yao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Mingxia Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Cixiong Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Changchuan Xie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yaying Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Han-Ming Wu
- Department of Neurology, First Affiliated Hospital, Xiamen University, Xiamen, China
| | - Li-Ping Fan
- Department of Neurology, First Affiliated Hospital, Xiamen University, Xiamen, China
| | - Jingxing Ou
- Department of Hepatic Surgery and Liver Transplantation Centre of the Third Affiliated Hospital, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China
| | - Yi-Hong Zhan
- Department of Neurology, First Affiliated Hospital, Xiamen University, Xiamen, China
| | - Shu-Yong Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China.
- Department of Digestive Diseases, School of Medicine, Xiamen University, Xiamen, China.
| | - Sheng-Cai Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
118
|
Di Maio G, Alessio N, Demirsoy IH, Peluso G, Perrotta S, Monda M, Di Bernardo G. Evaluation of Browning Agents on the White Adipogenesis of Bone Marrow Mesenchymal Stromal Cells: A Contribution to Fighting Obesity. Cells 2021; 10:403. [PMID: 33669222 PMCID: PMC7919793 DOI: 10.3390/cells10020403] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/28/2021] [Accepted: 02/10/2021] [Indexed: 01/19/2023] Open
Abstract
Brown-like adipocytes can be induced in white fat depots by a different environmental or drug stimuli, known as "browning" or "beiging". These brite adipocytes express thermogenin UCP1 protein and show different metabolic advantages, such as the ability to acquire a thermogenic phenotype corresponding to standard brown adipocytes that counteracts obesity. In this research, we evaluated the effects of several browning agents during white adipocyte differentiation of bone marrow-derived mesenchymal stromal cells (MSCs). Our in vitro findings identified two compounds that may warrant further in vivo investigation as possible anti-obesity drugs. We found that rosiglitazone and sildenafil are the most promising drug candidates for a browning treatment of obesity. These drugs are already available on the market for treating diabetes and erectile dysfunction, respectively. Thus, their off-label use may be contemplated, but it must be emphasized that some severe side effects are associated with use of these drugs.
Collapse
Affiliation(s)
- Girolamo Di Maio
- Human Physiology and Unit of Dietetic and Sports Medicine Section, Department of Experimental Medicine, School of Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (G.D.M.); (M.M.)
| | - Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, School of Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (N.A.); (I.H.D.)
| | - Ibrahim Halil Demirsoy
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, School of Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (N.A.); (I.H.D.)
| | | | - Silverio Perrotta
- Dipartimento della Donna, del Bambino e di Chirurgia Generale e Specialistica, School of Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy;
| | - Marcellino Monda
- Human Physiology and Unit of Dietetic and Sports Medicine Section, Department of Experimental Medicine, School of Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (G.D.M.); (M.M.)
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, School of Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (N.A.); (I.H.D.)
| |
Collapse
|
119
|
Li X, Wang L, Wang M, Zhang Z, Ma C, Ma X, Na X, Liang W. Global analysis of protein succinylation modification of Nostoc flagelliforme in response to dehydration. J Proteomics 2021; 237:104149. [PMID: 33588108 DOI: 10.1016/j.jprot.2021.104149] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/13/2021] [Accepted: 02/02/2021] [Indexed: 11/29/2022]
Abstract
Nostoc flagelliforme is a type of terrestrial cyanobacteria that is distributed in arid or semi-arid steppes in China. To research the molecular mechanisms underlying the adaptation of N. flagelliforme to drought stress, the succinylated expression profile and changes in N. flagelliforme that resulted as a response to dehydration were analyzed by label-free proteomics. A total of 1149 succinylated sites, 1128 succinylated peptides, and 396 succinylated proteins were identified. Succinylated proteins were differentially involved in photosynthesis and energy metabolism, as well as in reactive oxygen species (ROS) scavenging. Motif-X analysis of succinylated sites determined a succinylation motif [KxxG]. N. flagelliforme adapts to dehydration by increasing glucose metabolism and pentose phosphate pathway flux, and decreasing photosynthetic rate, which some of the key proteins were succinylated. ROS scavenging was mainly involved in the regulation of the enzyme antioxidant defense system and non-enzymatic antioxidant defense system through succinylation modification, thus eliminating excessive ROS. Protein succinylation of N. flagelliforme may play an important regulatory role in response to dehydration. The results are foundational, as they can inform future research into the mechanisms involved in the succinylation regulation mechanism of N. flagelliforme in response to dehydration. SIGNIFICANCE: The global succinylation network involved in response to dehydration in N. flagelliforme has been established. We found that many succinylated proteins were involved in photosynthesis, glucose metabolism and antioxidation. The global survey of succinylated proteins and the changes of succinylated levels in response to dehydration provided effective information for the drought tolerance mechanism in N. flagelliforme.
Collapse
Affiliation(s)
- Xiaoxu Li
- School of Life Sciences, Ningxia University, Yinchuan 750021, PR China
| | - Lingxia Wang
- School of Life Sciences, Ningxia University, Yinchuan 750021, PR China
| | - Meng Wang
- School of Life Sciences, Ningxia University, Yinchuan 750021, PR China
| | - Zheng Zhang
- School of Life Sciences, Ningxia University, Yinchuan 750021, PR China
| | - Caixia Ma
- School of Life Sciences, Ningxia University, Yinchuan 750021, PR China
| | - Xiaorong Ma
- School of Life Sciences, Ningxia University, Yinchuan 750021, PR China
| | - Xiaofan Na
- School of Life Sciences, Lanzhou University, Lanzhou 730000, PR China.
| | - Wenyu Liang
- School of Life Sciences, Ningxia University, Yinchuan 750021, PR China.
| |
Collapse
|
120
|
Senthivinayagam S, Serbulea V, Upchurch CM, Polanowska-Grabowska R, Mendu SK, Sahu S, Jayaguru P, Aylor KW, Chordia MD, Steinberg L, Oberholtzer N, Uchiyama S, Inada N, Lorenz UM, Harris TE, Keller SR, Meher AK, Kadl A, Desai BN, Kundu BK, Leitinger N. Adaptive thermogenesis in brown adipose tissue involves activation of pannexin-1 channels. Mol Metab 2021; 44:101130. [PMID: 33248294 PMCID: PMC7779784 DOI: 10.1016/j.molmet.2020.101130] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 11/06/2020] [Accepted: 11/21/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Brown adipose tissue (BAT) is specialized in thermogenesis. The conversion of energy into heat in brown adipocytes proceeds via stimulation of β-adrenergic receptor (βAR)-dependent signaling and activation of mitochondrial uncoupling protein 1 (UCP1). We have previously demonstrated a functional role for pannexin-1 (Panx1) channels in white adipose tissue; however, it is not known whether Panx1 channels play a role in the regulation of brown adipocyte function. Here, we tested the hypothesis that Panx1 channels are involved in brown adipocyte activation and thermogenesis. METHODS In an immortalized brown pre-adipocytes cell line, Panx1 currents were measured using patch-clamp electrophysiology. Flow cytometry was used for assessment of dye uptake and luminescence assays for adenosine triphosphate (ATP) release, and cellular temperature measurement was performed using a ratiometric fluorescence thermometer. We used RNA interference and expression plasmids to manipulate expression of wild-type and mutant Panx1. We used previously described adipocyte-specific Panx1 knockout mice (Panx1Adip-/-) and generated brown adipocyte-specific Panx1 knockout mice (Panx1BAT-/-) to study pharmacological or cold-induced thermogenesis. Glucose uptake into brown adipose tissue was quantified by positron emission tomography (PET) analysis of 18F-fluorodeoxyglucose (18F-FDG) content. BAT temperature was measured using an implantable telemetric temperature probe. RESULTS In brown adipocytes, Panx1 channel activity was induced either by apoptosis-dependent caspase activation or by β3AR stimulation via a novel mechanism that involves Gβγ subunit binding to Panx1. Inactivation of Panx1 channels in cultured brown adipocytes resulted in inhibition of β3AR-induced lipolysis, UCP-1 expression, and cellular thermogenesis. In mice, adiponectin-Cre-dependent genetic deletion of Panx1 in all adipose tissue depots resulted in defective β3AR agonist- or cold-induced thermogenesis in BAT and suppressed beigeing of white adipose tissue. UCP1-Cre-dependent Panx1 deletion specifically in brown adipocytes reduced the capacity for adaptive thermogenesis without affecting beigeing of white adipose tissue and aggravated diet-induced obesity and insulin resistance. CONCLUSIONS These data demonstrate that Gβγ-dependent Panx1 channel activation is involved in β3AR-induced thermogenic regulation in brown adipocytes. Identification of Panx1 channels in BAT as novel thermo-regulatory elements downstream of β3AR activation may have therapeutic implications.
Collapse
Affiliation(s)
| | - Vlad Serbulea
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Clint M Upchurch
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | | | - Suresh K Mendu
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Srabani Sahu
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Prathiba Jayaguru
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Kevin W Aylor
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Mahendra D Chordia
- Department of Radiology and Medical Imaging, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Limor Steinberg
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Nathaniel Oberholtzer
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Seichii Uchiyama
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Noriko Inada
- Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Ulrike M Lorenz
- Department of Microbiology, Immunology and Cancer Biology, Center for Cell Clearance, the Beirne B. Carter Center for Immunology Research, USA
| | - Thurl E Harris
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Susanna R Keller
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Akshaya K Meher
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Alexandra Kadl
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Bimal N Desai
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Bijoy K Kundu
- Department of Radiology and Medical Imaging, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA; Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA; Robert M Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
121
|
Quantification and Identification of Post-Translational Modifications Using Modern Proteomics Approaches. Methods Mol Biol 2021; 2228:225-235. [PMID: 33950494 PMCID: PMC8710235 DOI: 10.1007/978-1-0716-1024-4_16] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Post-translational modifications (PTMs) occur dynamically, allowing cells to quickly respond to changes in the environment. Lysine residues can be targeted by several modifications including acylations (acetylation, succinylation, malonylation, glutarylation, and others), methylation, ubiquitination, and other modifications. One of the most efficient methods for the identification of post-translational modifications is utilizing immunoaffinity enrichment followed by high-resolution mass spectrometry. This workflow can be coupled with comprehensive data-independent acquisition (DIA) mass spectrometry to be a high-throughput, label-free PTM quantification approach. Below we describe a detailed protocol to process tissue by homogenization and proteolytically digest proteins, followed by immunoaffinity enrichment of lysine-acetylated peptides to identify and quantify relative changes of acetylation comparing different conditions.
Collapse
|
122
|
Zhang Y, Otomaru K, Oshima K, Goto Y, Oshima I, Muroya S, Sano M, Roh S, Gotoh T. Maternal Nutrition During Gestation Alters Histochemical Properties, and mRNA and microRNA Expression in Adipose Tissue of Wagyu Fetuses. Front Endocrinol (Lausanne) 2021; 12:797680. [PMID: 35178028 PMCID: PMC8844027 DOI: 10.3389/fendo.2021.797680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022] Open
Abstract
We hypothesized that maternal low or high nutrition would give unique effects to morphological and molecular dynamics in adipose tissue of fetus of fatty breed Wagyu (Japanese Black) cattle which produce highly marbled beef. This study aimed to determine the effects of maternal energy intake in Wagyu cows, during gestation on fetal adipose tissue development, histochemical properties, and gene and microRNA (miRNA) expression. Cows were allocated to one of two nutritional energy groups: 120% (HIGH) or 60% nutritional requirements of (LOW). Fetuses (n = 6 per treatment) were removed from pregnant cows by cesarean section at fetal age 260 ± 8 days and euthanized. Subcutaneous adipose tissue (SAT), thoracic cavity visceral adipose tissue (TVAT), and perirenal adipose tissue (PAT) were collected for analysis. In histochemical analysis, in SAT and PAT, HIGH fetuses had greater diameter of adipocytes than LOW fetuses (P<0.05). Only in SAT, LOW fetuses had more Leptin (LEP) mRNA and tended to have more Peroxisome Proliferator-Activated Receptor gamma (PPARG) CCAAT-enhancer-binding proteins alpha (CEBPA) and Glucose transporter (GLUT) 4 mRNA(P<0.10). In all SAT, TVAT, and PAT, LOW fetuses had higher levels of the brown adipose tissue (BAT) biomarkers Uncoupling Protein (UCP) 1 and PPARG coactivator (PGC) 1α mRNA than HIGH fetuses (P<0.08). Meanwhile, in the other adipose tissue, LOW fetuses had lower PPARG, CEBPA, and Zinc Finger Protein (ZFP) 423 (in TVAT and PAT), FASN (in TVAT), LEP and GLUT4 mRNA (in PAT; P<0.10). In particular, in TVAT and PAT, LOW fetuses exhibited lower expression of WAT biomarkers (PPARG and ZFP423). Differential expression of various miRNAs related to adipogenesis between the LOW and HIGH fetuses was detected in an adipose tissue-specific manner (P<0.10). Based on adipose tissue-specific effects of maternal nutrition, these findings suggested that poor maternal nutrition in Wagyu cattle increased BAT development in SAT, TVAT and PAT, while elevated maternal nutrition stimulated fetal SAT development compared with that of TVAT and PAT.
Collapse
Affiliation(s)
- Yi Zhang
- Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
- Kuju Agricultural Research Center, Kyushu University, Taketa, Japan
| | - Konosuke Otomaru
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Kazunaga Oshima
- Western Region Agricultural Research Center, National Agriculture and Food Research Organization (NARO), Oda, Japan
| | - Yuji Goto
- Western Region Agricultural Research Center, National Agriculture and Food Research Organization (NARO), Oda, Japan
| | - Ichiro Oshima
- Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
| | - Susumu Muroya
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Tsukuba, Japan
| | - Mitsue Sano
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, Hikone, Japan
| | - Sanggun Roh
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Takafumi Gotoh
- Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
- Kuju Agricultural Research Center, Kyushu University, Taketa, Japan
- *Correspondence: Takafumi Gotoh,
| |
Collapse
|
123
|
Mu Q, Zuo J, Zhao D, Zhou X, Hua J, Bai Y, Mo F, Fang X, Fu M, Gao S. Ginsenoside rg3 reduces body weight by regulating fat content and browning in obese mice. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2021. [DOI: 10.1016/j.jtcms.2021.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
124
|
SUCLA2 mutations cause global protein succinylation contributing to the pathomechanism of a hereditary mitochondrial disease. Nat Commun 2020; 11:5927. [PMID: 33230181 PMCID: PMC7684291 DOI: 10.1038/s41467-020-19743-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial acyl-coenzyme A species are emerging as important sources of protein modification and damage. Succinyl-CoA ligase (SCL) deficiency causes a mitochondrial encephalomyopathy of unknown pathomechanism. Here, we show that succinyl-CoA accumulates in cells derived from patients with recessive mutations in the tricarboxylic acid cycle (TCA) gene succinyl-CoA ligase subunit-β (SUCLA2), causing global protein hyper-succinylation. Using mass spectrometry, we quantify nearly 1,000 protein succinylation sites on 366 proteins from patient-derived fibroblasts and myotubes. Interestingly, hyper-succinylated proteins are distributed across cellular compartments, and many are known targets of the (NAD+)-dependent desuccinylase SIRT5. To test the contribution of hyper-succinylation to disease progression, we develop a zebrafish model of the SCL deficiency and find that SIRT5 gain-of-function reduces global protein succinylation and improves survival. Thus, increased succinyl-CoA levels contribute to the pathology of SCL deficiency through post-translational modifications. The pathomechanism of succinyl-CoA ligase (SCL) deficiency, a hereditary mitochondrial disease, is not fully understood. Here, the authors show that increased succinyl-CoA levels contribute to SCL pathology by causing global protein hyper-succinylation.
Collapse
|
125
|
Helsley RN, Moreau F, Gupta MK, Radulescu A, DeBosch B, Softic S. Tissue-Specific Fructose Metabolism in Obesity and Diabetes. Curr Diab Rep 2020; 20:64. [PMID: 33057854 DOI: 10.1007/s11892-020-01342-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW The objective of this review is to provide up-to-date and comprehensive discussion of tissue-specific fructose metabolism in the context of diabetes, dyslipidemia, and nonalcoholic fatty liver disease (NAFLD). RECENT FINDINGS Increased intake of dietary fructose is a risk factor for a myriad of metabolic complications. Tissue-specific fructose metabolism has not been well delineated in terms of its contribution to detrimental health effects associated with fructose intake. Since inhibitors targeting fructose metabolism are being developed for the management of NAFLD and diabetes, it is essential to recognize how inability of one tissue to metabolize fructose may affect metabolism in the other tissues. The primary sites of fructose metabolism are the liver, intestine, and kidney. Skeletal muscle and adipose tissue can also metabolize a large portion of fructose load, especially in the setting of ketohexokinase deficiency, the rate-limiting enzyme of fructose metabolism. Fructose can also be sensed by the pancreas and the brain, where it can influence essential functions involved in energy homeostasis. Lastly, fructose is metabolized by the testes, red blood cells, and lens of the eye where it may contribute to infertility, advanced glycation end products, and cataracts, respectively. An increase in sugar intake, particularly fructose, has been associated with the development of obesity and its complications. Inhibition of fructose utilization in tissues primary responsible for its metabolism alters consumption in other tissues, which have not been traditionally regarded as important depots of fructose metabolism.
Collapse
Affiliation(s)
- Robert N Helsley
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Francois Moreau
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Manoj K Gupta
- Islet Cell and Regenerative Medicine, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Aurelia Radulescu
- Department of Pediatrics, University of Kentucky College of Medicine and Kentucky Children's Hospital, Lexington, KY, 40536, USA
| | - Brian DeBosch
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63131, USA
| | - Samir Softic
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, 40506, USA.
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, 138 Leader Ave, Lexington, KY, 40506, USA.
| |
Collapse
|
126
|
Wang Z, Ning T, Song A, Rutter J, Wang QA, Jiang L. Chronic cold exposure enhances glucose oxidation in brown adipose tissue. EMBO Rep 2020; 21:e50085. [PMID: 33043581 PMCID: PMC7645266 DOI: 10.15252/embr.202050085] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 09/02/2020] [Accepted: 09/10/2020] [Indexed: 01/20/2023] Open
Abstract
The cultured brown adipocytes can oxidize glucose in vitro, but it is still not fully clear whether brown adipose tissue (BAT) could completely oxidize glucose in vivo. Although positron emission tomography (PET) with 18F‐fluorodeoxyglucose (18F‐FDG) showed a high level of glucose uptake in the activated BAT, the non‐metabolizable 18F‐FDG cannot fully demonstrate intracellular glucose metabolism. Through in vivo [U‐13C]glucose tracing, here we show that chronic cold exposure dramatically activates glucose oxidation in BAT and the browning/beiging subcutaneous white adipose tissue (sWAT). Specifically, chronic cold exposure enhances glucose flux into the mitochondrial TCA cycle. Metabolic flux analysis models that β3‐adrenergic receptor (β3‐AR) agonist significantly enhances the flux of mitochondrial pyruvate uptake through mitochondrial pyruvate carrier (MPC) in the differentiated primary brown adipocytes. Furthermore, in vivo MPC inhibition blocks cold‐induced glucose oxidation and impairs body temperature maintenance in mice. Together, mitochondrial pyruvate uptake and oxidation serve an important energy source in the chronic cold exposure activated BAT and beige adipose tissue, which supports a role for glucose oxidation in brown fat thermogenesis.
Collapse
Affiliation(s)
- Zhichao Wang
- Department of Molecular & Cellular Endocrinology, Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Tinglu Ning
- Department of Molecular & Cellular Endocrinology, Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Anying Song
- Department of Molecular & Cellular Endocrinology, Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Jared Rutter
- Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT, USA.,Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Qiong A Wang
- Department of Molecular & Cellular Endocrinology, Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA.,Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Lei Jiang
- Department of Molecular & Cellular Endocrinology, Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA.,Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| |
Collapse
|
127
|
Xie N, Zhang L, Gao W, Huang C, Huber PE, Zhou X, Li C, Shen G, Zou B. NAD + metabolism: pathophysiologic mechanisms and therapeutic potential. Signal Transduct Target Ther 2020; 5:227. [PMID: 33028824 PMCID: PMC7539288 DOI: 10.1038/s41392-020-00311-7] [Citation(s) in RCA: 502] [Impact Index Per Article: 100.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/04/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) and its metabolites function as critical regulators to maintain physiologic processes, enabling the plastic cells to adapt to environmental changes including nutrient perturbation, genotoxic factors, circadian disorder, infection, inflammation and xenobiotics. These effects are mainly achieved by the driving effect of NAD+ on metabolic pathways as enzyme cofactors transferring hydrogen in oxidation-reduction reactions. Besides, multiple NAD+-dependent enzymes are involved in physiology either by post-synthesis chemical modification of DNA, RNA and proteins, or releasing second messenger cyclic ADP-ribose (cADPR) and NAADP+. Prolonged disequilibrium of NAD+ metabolism disturbs the physiological functions, resulting in diseases including metabolic diseases, cancer, aging and neurodegeneration disorder. In this review, we summarize recent advances in our understanding of the molecular mechanisms of NAD+-regulated physiological responses to stresses, the contribution of NAD+ deficiency to various diseases via manipulating cellular communication networks and the potential new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Na Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lu Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Wei Gao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Peter Ernst Huber
- CCU Molecular and Radiation Oncology, German Cancer Research Center; Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Xiaobo Zhou
- First Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Changlong Li
- West China School of Basic Medical Sciences & Forensic Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Guobo Shen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Bingwen Zou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
- CCU Molecular and Radiation Oncology, German Cancer Research Center; Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
- Department of Thoracic Oncology and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
128
|
Ruan HB. Developmental and functional heterogeneity of thermogenic adipose tissue. J Mol Cell Biol 2020; 12:775-784. [PMID: 32569352 PMCID: PMC7816678 DOI: 10.1093/jmcb/mjaa029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/11/2020] [Accepted: 06/12/2020] [Indexed: 12/18/2022] Open
Abstract
The obesity epidemic continues to rise as a global health challenge. Thermogenic brown and beige adipocytes dissipate chemical energy as heat, providing an opportunity for developing new therapeutics for obesity and related metabolic diseases. Anatomically, brown adipose tissue is distributed as discrete depots, while beige adipocytes exist within certain depots of white adipose tissue. Developmentally, brown and beige adipocytes arise from multiple embryonic progenitor populations that are distinct and overlapping. Functionally, they respond to a plethora of stimuli to engage uncoupling protein 1-dependent and independent thermogenic programs, thus improving systemic glucose homeostasis, lipid metabolism, and the clearance of branched-chain amino acids. In this review, we highlight recent advances in our understanding of the molecular and cellular mechanisms that contribute to the developmental and functional heterogeneity of thermogenic adipose tissue.
Collapse
Affiliation(s)
- Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
129
|
Ou T, Yang W, Li W, Lu Y, Dong Z, Zhu H, Sun X, Dong Z, Weng X, Chang S, Li H, Li Y, Qiu Z, Hu K, Sun A, Ge J. SIRT5 deficiency enhances the proliferative and therapeutic capacities of adipose-derived mesenchymal stem cells via metabolic switching. Clin Transl Med 2020; 10:e172. [PMID: 32997407 PMCID: PMC7510333 DOI: 10.1002/ctm2.172] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have therapeutic potential for multiple ischemic diseases. However, in vitro expansion of MSCs before clinical application leads to metabolic reprogramming from glycolysis to oxidative phosphorylation, drastically impairing their proliferative and therapeutic capacities. This study aimed to define the regulatory effects of Sirtuin 5 (SIRT5) on the proliferative and therapeutic functions of adipose-derived MSCs (ADMSCs) during in vitro expansion. METHODS ADMSCs were isolated from wild-type (WT) and Sirt5-knockout (Sirt5-/- ) mice. Cell counting assay was used to investigate the proliferative capacities of the ADMSCs. Dihydroethidium and senescence-associated β-galactosidase stainings were used to measure intracellular ROS and senescence levels. Mass spectrometry was used to analyze protein succinylation. Oxygen consumption rates and extra cellular acidification rates were measured as indicators of mitochondrial respiration and glycolysis. Metabolic-related genes expression were verified by quantitative PCR and western blot. Hind limb ischemia mouse model was used to evaluate the therapeutic potentials of WT and Sirt5-/- ADSMCs. RESULTS SIRT5 protein levels were upregulated in ADMCs during in vitro expansion. Sirt5-/- ADMSCs exhibited a higher proliferation rate, delayed senescence, and reduced ROS accumulation. Furthermore, elevated protein succinylation levels were observed in Sirt5-/- ADMSCs, leading to the reduced activity of tricarboxylic acid cycle-related enzymes and attenuated mitochondrial respiration. Glucose uptake, glycolysis, and pentose phosphate pathway were elevated in Sirt5-/- ADMSCs. Inhibition of succinylation by glycine or re-expression of Sirt5 reversed the metabolic alterations in Sirt5-/- ADMSCs, thus abolishing their enhanced proliferative capacities. In the hind limb ischemia mouse model, SIRT5-/- ADMSCs transplantation enhanced blood flow recovery and angiogenesis compared with WT ADMSCs. CONCLUSIONS Our results indicate that SIRT5 deficiency during ADMSC culture expansion leads to reversed metabolic pattern, enhanced proliferative capacities, and improved therapeutic outcomes. These data suggest SIRT5 as a potential target to enhance the functional properties of MSCs for clinical application.
Collapse
Affiliation(s)
- Tiantong Ou
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Wenlong Yang
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Wenjia Li
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Yijing Lu
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
| | - Zheng Dong
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Hongming Zhu
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Xiaolei Sun
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Zhen Dong
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Xinyu Weng
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Suchi Chang
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Hua Li
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Yufan Li
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Zhiwei Qiu
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Kai Hu
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
| | - Aijun Sun
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
| | - Junbo Ge
- Department of Cardiology, Zhongshan HospitalFudan University, Shanghai Institute of Cardiovascular DiseasesShanghaiChina
- Institute of Biomedical SciencesFudan UniversityShanghaiChina
| |
Collapse
|
130
|
Li X, Zhang C, Zhao T, Su Z, Li M, Hu J, Wen J, Shen J, Wang C, Pan J, Mu X, Ling T, Li Y, Wen H, Zhang X, You Q. Lysine-222 succinylation reduces lysosomal degradation of lactate dehydrogenase a and is increased in gastric cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:172. [PMID: 32859246 PMCID: PMC7455916 DOI: 10.1186/s13046-020-01681-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 08/17/2020] [Indexed: 01/07/2023]
Abstract
Background Lysine succinylation is an emerging posttranslational modification that has garnered increased attention recently, but its role in gastric cancer (GC) remains underexplored. Methods Proteomic quantification of lysine succinylation was performed in human GC tissues and adjacent normal tissues by mass spectrometry. The mRNA and protein levels of lactate dehydrogenase A (LDHA) in GC and adjacent normal tissues were analyzed by qRT-PCR and western blot, respectively. The expression of K222-succinylated LDHA was measured in GC tissue microarray by the K222 succinylation-specific antibody. The interaction between LDHA and sequestosome 1 (SQSTM1) was measured by co-immunoprecipitation (co-IP) and proximity ligation assay (PLA). The binding of carnitine palmitoyltransferase 1A (CPT1A) to LDHA was determined by co-IP. The effect of K222-succinylated LDHA on tumor growth and metastasis was evaluated by in vitro and in vivo experiments. Results Altogether, 503 lysine succinylation sites in 303 proteins were identified. Lactate dehydrogenase A (LDHA), the key enzyme in Warburg effect, was found highly succinylated at K222 in GC. Intriguingly, this modification did not affect LDHA ubiquitination, but reduced the binding of ubiquitinated LDHA to SQSTM1, thereby decreasing its lysosomal degradation. We demonstrated that CPT1A functions as a lysine succinyltransferase that interacts with and succinylates LDHA. Moreover, high K222-succinylation of LDHA was associated with poor prognosis in patients with GC. Finally, overexpression of a succinylation-mimic mutant of LDHA promoted cell proliferation, invasion, and migration. Conclusions Our data revealed a novel lysosomal pathway of LDHA degradation, which is mediated by the binding of K63-ubiquitinated LDHA to SQSTM1. Strikingly, CPT1A succinylates LDHA on K222, which thereby reduces the binding and inhibits the degradation of LDHA, as well as promotes GC invasion and proliferation. This study thus uncovers a new role of lysine succinylation and the mechanism underlying LDHA upregulation in GC.
Collapse
Affiliation(s)
- Xiang Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Department of Biotherapy, Department of Surgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China.,Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Zhang
- Department of Biotherapy, Department of Surgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Ting Zhao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Zhongping Su
- Department of Biotherapy, Department of Surgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Mengjing Li
- Department of Biotherapy, Department of Surgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Jiancheng Hu
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, 169610, Singapore.,Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Jianfei Wen
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jiajia Shen
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chao Wang
- Department of Biotherapy, Department of Surgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Jinshun Pan
- Department of Biotherapy, Department of Surgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Xianmin Mu
- Department of Biotherapy, Department of Surgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Tao Ling
- Department of Biotherapy, Department of Surgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Yingchang Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Hao Wen
- Department of Biotherapy, Department of Surgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Xiaoren Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou, 510182, China
| | - Qiang You
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China. .,Department of Biotherapy, Department of Surgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China. .,Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou, 510182, China.
| |
Collapse
|
131
|
Trefely S, Lovell CD, Snyder NW, Wellen KE. Compartmentalised acyl-CoA metabolism and roles in chromatin regulation. Mol Metab 2020; 38:100941. [PMID: 32199817 PMCID: PMC7300382 DOI: 10.1016/j.molmet.2020.01.005] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/03/2020] [Accepted: 01/07/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Many metabolites serve as important signalling molecules to adjust cellular activities and functions based on nutrient availability. Links between acetyl-CoA metabolism, histone lysine acetylation, and gene expression have been documented and studied over the past decade. In recent years, several additional acyl modifications to histone lysine residues have been identified, which depend on acyl-coenzyme A thioesters (acyl-CoAs) as acyl donors. Acyl-CoAs are intermediates of multiple distinct metabolic pathways, and substantial evidence has emerged that histone acylation is metabolically sensitive. Nevertheless, the metabolic sources of acyl-CoAs used for chromatin modification in most cases remain poorly understood. Elucidating how these diverse chemical modifications are coupled to and regulated by cellular metabolism is important in deciphering their functional significance. SCOPE OF REVIEW In this article, we review the metabolic pathways that produce acyl-CoAs, as well as emerging evidence for functional roles of diverse acyl-CoAs in chromatin regulation. Because acetyl-CoA has been extensively reviewed elsewhere, we will focus on four other acyl-CoA metabolites integral to major metabolic pathways that are also known to modify histones: succinyl-CoA, propionyl-CoA, crotonoyl-CoA, and butyryl-CoA. We also briefly mention several other acyl-CoA species, which present opportunities for further research; malonyl-CoA, glutaryl-CoA, 3-hydroxybutyryl-CoA, 2-hydroxyisobutyryl-CoA, and lactyl-CoA. Each acyl-CoA species has distinct roles in metabolism, indicating the potential to report shifts in the metabolic status of the cell. For each metabolite, we consider the metabolic pathways in which it participates and the nutrient sources from which it is derived, the compartmentalisation of its metabolism, and the factors reported to influence its abundance and potential nuclear availability. We also highlight reported biological functions of these metabolically-linked acylation marks. Finally, we aim to illuminate key questions in acyl-CoA metabolism as they relate to the control of chromatin modification. MAJOR CONCLUSIONS A majority of acyl-CoA species are annotated to mitochondrial metabolic processes. Since acyl-CoAs are not known to be directly transported across mitochondrial membranes, they must be synthesized outside of mitochondria and potentially within the nucleus to participate in chromatin regulation. Thus, subcellular metabolic compartmentalisation likely plays a key role in the regulation of histone acylation. Metabolite tracing in combination with targeting of relevant enzymes and transporters will help to map the metabolic pathways that connect acyl-CoA metabolism to chromatin modification. The specific function of each acyl-CoA may be determined in part by biochemical properties that affect its propensity for enzymatic versus non-enzymatic protein modification, as well as the various enzymes that can add, remove and bind each modification. Further, competitive and inhibitory effects of different acyl-CoA species on these enzymes make determining the relative abundance of acyl-CoA species in specific contexts important to understand the regulation of chromatin acylation. An improved and more nuanced understanding of metabolic regulation of chromatin and its roles in physiological and disease-related processes will emerge as these questions are answered.
Collapse
Affiliation(s)
- Sophie Trefely
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Metabolic Disease Research, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Claudia D Lovell
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nathaniel W Snyder
- Center for Metabolic Disease Research, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
132
|
Roles of Mitochondrial Sirtuins in Mitochondrial Function, Redox Homeostasis, Insulin Resistance and Type 2 Diabetes. Int J Mol Sci 2020; 21:ijms21155266. [PMID: 32722262 PMCID: PMC7432223 DOI: 10.3390/ijms21155266] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are the metabolic hubs that process a number of reactions including tricarboxylic acid cycle, β-oxidation of fatty acids and part of the urea cycle and pyrimidine nucleotide biosynthesis. Mitochondrial dysfunction impairs redox homeostasis and metabolic adaptation, leading to aging and metabolic disorders like insulin resistance and type 2 diabetes. SIRT3, SIRT4 and SIRT5 belong to the sirtuin family proteins and are located at mitochondria and also known as mitochondrial sirtuins. They catalyze NAD+-dependent deacylation (deacetylation, demalonylation and desuccinylation) and ADP-ribosylation and modulate the function of mitochondrial targets to regulate the metabolic status in mammalian cells. Emerging evidence has revealed that mitochondrial sirtuins coordinate the regulation of gene expression and activities of a wide spectrum of enzymes to orchestrate oxidative metabolism and stress responses. Mitochondrial sirtuins act in synergistic or antagonistic manners to promote respiratory function, antioxidant defense, insulin response and adipogenesis to protect individuals from aging and aging-related metabolic abnormalities. In this review, we focus on the molecular mechanisms by which mitochondrial sirtuins regulate oxidative metabolism and antioxidant defense and discuss the roles of their deficiency in the impairment of mitochondrial function and pathogenesis of insulin resistance and type 2 diabetes.
Collapse
|
133
|
Yao B, Li Y, Niu Y, Wang L, Chen T, Guo C, Liu Q. Hypoxia-induced miR-3677-3p promotes the proliferation, migration and invasion of hepatocellular carcinoma cells by suppressing SIRT5. J Cell Mol Med 2020; 24:8718-8731. [PMID: 32596968 PMCID: PMC7412699 DOI: 10.1111/jcmm.15503] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 05/08/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC), with life‐threatening malignant behaviours, often develops distant metastases and is the fourth most common primary cancer in the world, having taken millions of lives in Asian countries such as China. The novel miR‐3677‐3p is involved in a high‐expression‐related poor prognosis in HCC tissues and cell lines, indicating oncogenesis functions in vitro and in vivo. Initially, we confirmed the inhibition of proliferation, migration and invasion in miR‐3677‐3p knock‐down MHCC‐97H and SMMC‐7721 cell lines, which are well known for their high degree of invasiveness. Then, we reversed the functional experiments in the low‐miR‐3677‐3p‐expression Hep3B cell line via overexpressing miR‐3677‐3p. In nude mice xenograft and lung metastasis assays, we found suppressor behaviours, smaller nodules and low density of organ spread, after injection of cells transfected with shRNA‐miR‐3677‐3p. A combination of databases (Starbase, TargetScan and MiRgator) illustrated miR‐3677‐3p targets, and it was shown to suppress the expression of SIRT5 in a dual‐luciferase reporter system. To clarify the conclusions of previous ambiguous research, we up‐regulated SIRT5 in Hep3B cells, and rescue tests were established for confirmation that miR‐3677‐3p suppresses SIRT5 to enhance the migration and invasion of HCC. Interestingly, we discovered hypoxia‐induced miR‐3677‐3p up‐regulation benefited HCC malignancy and invasiveness. In conclusion, the overexpression of miR‐3677‐3p mediated SIRT5 inhibition, which could increase proliferation, migration and invasion of HCC in hypoxic microenvironments.
Collapse
Affiliation(s)
- Bowen Yao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yazhao Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yongshen Niu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tianxiang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Cheng Guo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qingguang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
134
|
The Role of Pref-1 during Adipogenic Differentiation: An Overview of Suggested Mechanisms. Int J Mol Sci 2020; 21:ijms21114104. [PMID: 32526833 PMCID: PMC7312882 DOI: 10.3390/ijms21114104] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 12/15/2022] Open
Abstract
Obesity contributes significantly to the global health burden. A better understanding of adipogenesis, the process of fat formation, may lead to the discovery of novel treatment strategies. However, it is of concern that the regulation of adipocyte differentiation has predominantly been studied using the murine 3T3-L1 preadipocyte cell line and murine experimental animal models. Translation of these findings to the human setting requires confirmation using experimental models of human origin. The ability of mesenchymal stromal/stem cells (MSCs) to differentiate into adipocytes is an attractive model to study adipogenesis in vitro. Differences in the ability of MSCs isolated from different sources to undergo adipogenic differentiation, may be useful in investigating elements responsible for regulating adipogenic differentiation potential. Genes involved may be divided into three broad categories: early, intermediate and late-stage regulators. Preadipocyte factor-1 (Pref-1) is an early negative regulator of adipogenic differentiation. In this review, we briefly discuss the adipogenic differentiation potential of MSCs derived from two different sources, namely adipose-derived stromal/stem cells (ASCs) and Wharton’s Jelly derived stromal/stem cells (WJSCs). We then discuss the function and suggested mechanisms of action of Pref-1 in regulating adipogenesis, as well as current findings regarding Pref-1’s role in human adipogenesis.
Collapse
|
135
|
Zhang C, He X, Sheng Y, Xu J, Yang C, Zheng S, Liu J, Li H, Ge J, Yang M, Zhai B, Xu W, Luo Y, Huang K. Allicin Regulates Energy Homeostasis through Brown Adipose Tissue. iScience 2020; 23:101113. [PMID: 32413611 PMCID: PMC7226876 DOI: 10.1016/j.isci.2020.101113] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/15/2020] [Accepted: 04/24/2020] [Indexed: 12/19/2022] Open
Abstract
Brown adipose tissue (BAT) is a promising potential therapeutic target for the treatment of obesity and related metabolic diseases. Allicin, a natural product in garlic, has multiple biological and pharmacological functions. However, the role of allicin in the regulation of metabolic organs, particularly BAT activation, has not been well studied. Here, we show that allicin imparts a significant effect by inhibiting body weight gain, decreasing adiposity, maintaining glucose homeostasis, improving insulin resistance, and ameliorating hepatic steatosis in obese mice. These observations strongly correlate with the activation of BAT. Notably, allicin plays a role in BAT activation, which may partly contribute to the Sirt1-PGC1α-Tfam pathway. In addition, allicin can significantly increase the succinylation levels of UCP1 in BAT by inhibiting sirt5, whereas excess allicin induces autophagy/mitophagy and mitochondrial dysfunction. Thus, our findings point to allicin as a promising therapeutic approach for the treatment of obesity and metabolic disorders. Allicin reduces adiposity and maintains glucose homeostasis Allicin activates the brown adipocytes and increases the energy expenditure Allicin enhances BAT activity partly through SIRT1-PGC1a-Tfam signaling pathway Allicin regulates mitophagy via suppressed sirt5-mediated succinylation accumulation
Collapse
Affiliation(s)
- Chuanhai Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Xiaoyun He
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Yao Sheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Jia Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Cui Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Shujuan Zheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Junyu Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Haoyu Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Jianbing Ge
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Minglan Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Baiqiang Zhai
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Wentao Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Yunbo Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China.
| | - Kunlun Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China.
| |
Collapse
|
136
|
Hussain MF, Roesler A, Kazak L. Regulation of adipocyte thermogenesis: mechanisms controlling obesity. FEBS J 2020; 287:3370-3385. [PMID: 32301220 DOI: 10.1111/febs.15331] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/26/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Adipocyte biology has been intensely researched in recent years due to the emergence of obesity as a serious global health concern and because of the realization that adipose tissue is more than simply a cell type that stores and releases lipids. The plasticity of adipose tissues, to rapidly adapt to altered physiological states of energy demand, is under neuronal and endocrine control. The capacity for white adipocytes to store chemical energy in lipid droplets is key for protecting other organs from the toxic effects of ectopic lipid deposition. In contrast, thermogenic (brown and beige) adipocytes combust macronutrients to generate heat. The thermogenic activity of adipocytes allows them to protect themselves and other tissues from lipid overaccumulation. Advances in brown fat biology have uncovered key molecular players involved in adipocyte determination, differentiation, and thermogenic activation. It is now, well appreciated that three distinct adipocyte types exist: white, beige, and brown. Moreover, functional differences are present within adipocyte subtypes located in anatomically distinct locations. Adding to this complexity is the recent realization from single-cell sequencing studies that adipocyte progenitors are also heterogeneous. Understanding the molecular details of how to increase the number of thermogenic fat cells and their activation may delineate some of the pathophysiological basis of obesity and obesity-related diseases. Here, we review recent advances that have extended our understanding of the central role that adipose tissue plays in energy balance and the mechanisms that control their amount and function.
Collapse
Affiliation(s)
- Mohammed Faiz Hussain
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Anna Roesler
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Lawrence Kazak
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| |
Collapse
|
137
|
Cruz JV, Maba IK, Correia D, Kaziuk FD, Cadena SMSC, Zampronio AR. Intermittent binge-like ethanol exposure during adolescence attenuates the febrile response by reducing brown adipose tissue thermogenesis in rats. Drug Alcohol Depend 2020; 209:107904. [PMID: 32078977 DOI: 10.1016/j.drugalcdep.2020.107904] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 01/28/2023]
Abstract
Ethanol (EtOH) consumption is a primary health risk worldwide, which generally starts during adolescence in a binge pattern (i.e., the episodic consumption of high amounts). Binge EtOH consumption can lead to modifications of the innate and adaptive immune responses, including fever. The present study evaluated the febrile response that was induced by lipopolysaccharide (LPS) and prostaglandins E2 (PGE2) and the mechanisms of thermoregulation in adolescent rats that were exposed to EtOH in a binge-like pattern. Male Wistar rats were treated with an intraperitoneal (i.p.) injection of EtOH or saline on postnatal days (PND) 25, 26, 29, 30, 33, 34, 37, and 38. On PND 51, they received a pyrogenic challenge with LPS (i.p.) or PGE2 (intracerebroventricular) to induce a febrile response. Interscapular brown adipose tissue (BAT) mass and uncoupling protein (UCP) activity in isolated mitochondria were evaluated on PND 51. The rats were then subjected to cold challenges to analyze adaptive thermogenesis. Intermittent EtOH exposure during adolescence impaired the LPS- and PGE2-induced febrile response 12 days after the end of EtOH exposure. Ethanol exposure decreased interscapular BAT mass, oxygen consumption, and UCP activity in isolated mitochondria, resulting in an impairment in thermogenesis at 5 °C. No morphological changes in BAT were observed. These findings indicate that binge-like EtOH exposure during adolescence impairs thermoregulation by reducing BAT mass and function. This reduction may last for a prolonged period of time after the cessation of EtOH exposure and may affect both cold defenses and the febrile response during the development of infectious diseases.
Collapse
Affiliation(s)
- J V Cruz
- Department of Pharmacology, Federal University of Paraná, PO Box 19031, 81531-980, Curitiba, PR, Brazil
| | - I K Maba
- Department of Pharmacology, Federal University of Paraná, PO Box 19031, 81531-980, Curitiba, PR, Brazil
| | - D Correia
- Department of Pharmacology, Federal University of Paraná, PO Box 19031, 81531-980, Curitiba, PR, Brazil
| | - F D Kaziuk
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, PO Box 19031, 81531-980, Curitiba, PR, Brazil
| | - S M S C Cadena
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, PO Box 19031, 81531-980, Curitiba, PR, Brazil
| | - A R Zampronio
- Department of Pharmacology, Federal University of Paraná, PO Box 19031, 81531-980, Curitiba, PR, Brazil.
| |
Collapse
|
138
|
Comito G, Ippolito L, Chiarugi P, Cirri P. Nutritional Exchanges Within Tumor Microenvironment: Impact for Cancer Aggressiveness. Front Oncol 2020; 10:396. [PMID: 32266157 PMCID: PMC7105815 DOI: 10.3389/fonc.2020.00396] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
Neoplastic tissues are composed not only by tumor cells but also by several non-transformed stromal cells, such as cancer-associated fibroblasts, endothelial and immune cells, that actively participate to tumor progression. Starting from the very beginning of carcinogenesis, tumor cells, through the release of paracrine soluble factors and vesicles, i.e., exosomes, modify the behavior of the neighboring cells, so that they can give efficient support for cancer cell proliferation and spreading. A mandatory role in tumor progression has been recently acknowledged to metabolic deregulation. Beside undergoing a metabolic reprogramming coherent to their high proliferation rate, tumor cells also rewire the metabolic assets of their stromal cells, educating them to serve as nutrient donors. Hence, an alteration in the composition and in the flow rate of many nutrients within tumor microenvironment has been associated with malignancy progression. This review is focused on metabolic remodeling of the different cell populations within tumor microenvironment, dealing with reciprocal re-education through the symbiotic sharing of metabolites, behaving both as nutrients and as transcriptional regulators, describing their impact on tumor growth and metastasis.
Collapse
Affiliation(s)
- Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.,Excellence Center for Research, Transfer and High Education DenoTHE, University of Florence, Florence, Italy
| | - Paolo Cirri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.,Excellence Center for Research, Transfer and High Education DenoTHE, University of Florence, Florence, Italy
| |
Collapse
|
139
|
Xie X, Shah S, Holtz A, Rose J, Basisty N, Schilling B. Simultaneous Affinity Enrichment of Two Post-Translational Modifications for Quantification and Site Localization. J Vis Exp 2020:10.3791/60780. [PMID: 32176209 PMCID: PMC7275731 DOI: 10.3791/60780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Studying multiple post-translational modifications (PTMs) of proteins is a crucial step to understand PTM crosstalk and gain more holistic insights into protein function. Despite the importance of multi-PTM enrichment studies, few studies investigate more than one PTM at a time, due partially to the expenses, time, and large protein quantities required to perform multiple global proteomic analysis of PTMs. The "one-pot" affinity enrichment detailed in this protocol overcomes these barriers by permitting the simultaneous identification and quantification of peptides with lysine residues containing acetylation and succinylation PTMs with low amounts of sample input. The protocol involves preparation of protein lysate from mouse livers of SIRT5 knockout mice, performance of trypsin digestion, enrichment for PTMs, and performance of mass spectrometric analysis using a data-independent acquisition (DIA) workflow. Because this workflow allows for the enrichment of two PTMs from the same sample simultaneously, it provides a practical tool to study PTM crosstalk without requiring large amounts of samples, and it greatly reduces the time required for sample preparation, data acquisition, and analysis. The DIA component of the workflow provides comprehensive PTM-specific information. This is particularly important when studying PTM site localization, as DIA provides comprehensive sets of fragment ions that can be computationally deciphered to differentiate between different PTM localization isoforms.
Collapse
|
140
|
Ebert SM, Bullard SA, Basisty N, Marcotte GR, Skopec ZP, Dierdorff JM, Al-Zougbi A, Tomcheck KC, DeLau AD, Rathmacher JA, Bodine SC, Schilling B, Adams CM. Activating transcription factor 4 (ATF4) promotes skeletal muscle atrophy by forming a heterodimer with the transcriptional regulator C/EBPβ. J Biol Chem 2020; 295:2787-2803. [PMID: 31953319 PMCID: PMC7049960 DOI: 10.1074/jbc.ra119.012095] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/10/2020] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle atrophy is a highly-prevalent and debilitating condition that remains poorly understood at the molecular level. Previous work found that aging, fasting, and immobilization promote skeletal muscle atrophy via expression of activating transcription factor 4 (ATF4) in skeletal muscle fibers. However, the direct biochemical mechanism by which ATF4 promotes muscle atrophy is unknown. ATF4 is a member of the basic leucine zipper transcription factor (bZIP) superfamily. Because bZIP transcription factors are obligate dimers, and because ATF4 is unable to form highly-stable homodimers, we hypothesized that ATF4 may promote muscle atrophy by forming a heterodimer with another bZIP family member. To test this hypothesis, we biochemically isolated skeletal muscle proteins that associate with the dimerization- and DNA-binding domain of ATF4 (the bZIP domain) in mouse skeletal muscle fibers in vivo Interestingly, we found that ATF4 forms at least five distinct heterodimeric bZIP transcription factors in skeletal muscle fibers. Furthermore, one of these heterodimers, composed of ATF4 and CCAAT enhancer-binding protein β (C/EBPβ), mediates muscle atrophy. Within skeletal muscle fibers, the ATF4-C/EBPβ heterodimer interacts with a previously unrecognized and evolutionarily conserved ATF-C/EBP composite site in exon 4 of the Gadd45a gene. This three-way interaction between ATF4, C/EBPβ, and the ATF-C/EBP composite site activates the Gadd45a gene, which encodes a critical mediator of muscle atrophy. Together, these results identify a biochemical mechanism by which ATF4 induces skeletal muscle atrophy, providing molecular-level insights into the etiology of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Scott M Ebert
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246; Emmyon, Inc., Coralville, Iowa 52241
| | - Steven A Bullard
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Nathan Basisty
- Buck Institute for Research on Aging, Novato, California 94945
| | - George R Marcotte
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Zachary P Skopec
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Jason M Dierdorff
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Asma Al-Zougbi
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Kristin C Tomcheck
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Austin D DeLau
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Jacob A Rathmacher
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Sue C Bodine
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Emmyon, Inc., Coralville, Iowa 52241
| | | | - Christopher M Adams
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246; Emmyon, Inc., Coralville, Iowa 52241.
| |
Collapse
|
141
|
Yan D, Franzini A, Pomicter AD, Halverson BJ, Antelope O, Mason CC, Ahmann JM, Senina AV, Vellore NA, Jones CL, Zabriskie MS, Than H, Xiao MJ, van Scoyk A, Patel AB, Clair PM, Heaton WL, Owen SC, Andersen JL, Egbert CM, Reisz JA, D'Alessandro A, Cox JE, Gantz KC, Redwine HM, Iyer SM, Khorashad JS, Rajabi N, Olsen CA, O'Hare T, Deininger MW. SIRT5 IS A DRUGGABLE METABOLIC VULNERABILITY IN ACUTE MYELOID LEUKEMIA. Blood Cancer Discov 2019; 2:266-287. [PMID: 34027418 DOI: 10.1158/2643-3230.bcd-20-0168] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
We discovered that the survival and growth of many primary acute myeloid leukemia (AML) samples and cell lines, but not normal CD34+ cells, are dependent on SIRT5, a lysine deacylase implicated in regulating multiple metabolic pathways. Dependence on SIRT5 is genotype-agnostic and extends to RAS- and p53-mutated AML. Results were comparable between SIRT5 knockdown and SIRT5 inhibition using NRD167, a potent and selective SIRT5 inhibitor. Apoptosis induced by SIRT5 disruption is preceded by reductions in oxidative phosphorylation and glutamine utilization, and an increase in mitochondrial superoxide that is attenuated by ectopic superoxide dismutase 2. These data indicate that SIRT5 controls and coordinates several key metabolic pathways in AML and implicate SIRT5 as a vulnerability in AML.
Collapse
Affiliation(s)
- Dongqing Yan
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Anca Franzini
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | | | - Orlando Antelope
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Clinton C Mason
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Jonathan M Ahmann
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Anna V Senina
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nadeem A Vellore
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Courtney L Jones
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Hein Than
- Department of Haematology, Singapore General Hospital, Singapore
| | - Michael J Xiao
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Ami B Patel
- Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT, USA
| | - Phillip M Clair
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - William L Heaton
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Shawn C Owen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
| | - Joshua L Andersen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Christina M Egbert
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Angelo D'Alessandro
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - James E Cox
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Kevin C Gantz
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Hannah M Redwine
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Siddharth M Iyer
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Jamshid S Khorashad
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Nima Rajabi
- Center for Biopharmaceuticals & Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Christian A Olsen
- Center for Biopharmaceuticals & Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Thomas O'Hare
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT, USA
| | - Michael W Deininger
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
142
|
Abstract
PURPOSE OF REVIEW Obesity is a major risk factor for type 2 diabetes. Although adipose tissue allows storage of excess calories in periods of overnutrition, in obesity, adipose tissue metabolism becomes dysregulated and can promote metabolic diseases. This review discusses recent advances in understandings how adipocyte metabolism impacts metabolic homeostasis. RECENT FINDINGS The ability of adipocytes to synthesize lipids from glucose is a marker of metabolic fitness, e.g., low de novo lipogenesis (DNL) in adipocytes correlates with insulin resistance in obesity. Adipocyte DNL may promote synthesis of special "insulin sensitizing" signaling lipids that act hormonally. However, each metabolic intermediate in the DNL pathway (i.e., citrate, acetyl-CoA, malonyl-CoA, and palmitate) also has second messenger functions. Mounting evidence suggests these signaling functions may also be important for maintaining healthy adipocytes. While adipocyte DNL contributes to lipid storage, lipid precursors may have additional second messenger functions critical for maintaining adipocyte health, and thus systemic metabolic homeostasis.
Collapse
Affiliation(s)
- Wen-Yu Hsiao
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA, 01605, USA
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
143
|
Dichotomous Sirtuins: Implications for Drug Discovery in Neurodegenerative and Cardiometabolic Diseases. Trends Pharmacol Sci 2019; 40:1021-1039. [PMID: 31704173 DOI: 10.1016/j.tips.2019.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/12/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023]
Abstract
Sirtuins (SIRT1-7), a class of NAD+-dependent deacylases, are central regulators of metabolic homeostasis and stress responses. While numerous salutary effects associated with sirtuin activation, especially SIRT1, are well documented, other reports show health benefits resulting from sirtuin inhibition. Furthermore, conflicting findings have been obtained regarding the pathophysiological role of specific sirtuin isoforms, suggesting that sirtuins act as 'double-edged swords'. Here, we provide an integrated overview of the different findings on the role of mammalian sirtuins in neurodegenerative and cardiometabolic disorders and attempt to dissect the reasons behind these different effects. Finally, we discuss how addressing these obstacles may provide a better understanding of the complex sirtuin biology and improve the likelihood of identifying effective and selective drug targets for a variety of human disorders.
Collapse
|