A newly identified compound activating UCP1 inhibits obesity and its related metabolic disorders
Ken Onodera
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorCorresponding Author
Yutaka Hasegawa
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Correspondence Yutaka Hasegawa, Iwate Medical University, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
Email: [email protected]
Search for more papers by this authorNozomi Yokota
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorShukuko Tamura
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorHirofumi Kinno
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorIwao Takahashi
Division of Molecular and Cellular Pharmacology, Department of Pathophysiology and Pharmacology, School of Pharmacy, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorHiraku Chiba
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorHirotatsu Kojima
Drug Discovery Initiative, The University of Tokyo, Tokyo, Japan
Search for more papers by this authorHideki Katagiri
Department of Diabetes and Metabolism, Tohoku University Graduate School of Medicine, Tohoku University Hospital, Sendai, Japan
Search for more papers by this authorKoji Nata
Division of Medical Biochemistry, School of Pharmacy, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorYasushi Ishigaki
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorKen Onodera
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorCorresponding Author
Yutaka Hasegawa
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Correspondence Yutaka Hasegawa, Iwate Medical University, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
Email: [email protected]
Search for more papers by this authorNozomi Yokota
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorShukuko Tamura
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorHirofumi Kinno
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorIwao Takahashi
Division of Molecular and Cellular Pharmacology, Department of Pathophysiology and Pharmacology, School of Pharmacy, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorHiraku Chiba
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorHirotatsu Kojima
Drug Discovery Initiative, The University of Tokyo, Tokyo, Japan
Search for more papers by this authorHideki Katagiri
Department of Diabetes and Metabolism, Tohoku University Graduate School of Medicine, Tohoku University Hospital, Sendai, Japan
Search for more papers by this authorKoji Nata
Division of Medical Biochemistry, School of Pharmacy, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorYasushi Ishigaki
Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
Search for more papers by this authorKen Onodera and Yutaka Hasegawa contributed equally to this work.
Abstract
Objective
Promoting thermogenesis in adipose tissue has been a promising strategy against obesity and related metabolic complications. We aimed to identify compounds that promote thermogenesis in adipocytes and to elucidate their functions and roles in metabolism.
Methods
To identify compounds that directly promote thermogenesis from a structurally diverse set of 4800 compounds, we utilized a cell-based platform for high-throughput screening that induces uncoupling protein 1 (Ucp1) expression in adipocytes.
Results
We identified one candidate compound that activates UCP1. Additional characterization of this compound revealed that it induced cellular thermogenesis in adipocytes with negligible cytotoxicity. In a subsequent diet-induced obesity model, mice treated with this compound exhibited a slower rate of weight gain, improved insulin sensitivity, and increased energy expenditure. Mechanistic studies have revealed that this compound increases mitochondrial biogenesis by elevating maximal respiration, which is partly mediated by the protein kinase A (PKA)-p38 mitogen-activated protein kinase (MAPK) signaling pathway. A further comprehensive genetic analysis of adipocytes treated with these compounds identified two novel UCP1-dependent thermogenic genes, potassium voltage-gated channel subfamily C member 2 (Kcnc2) and predicted gene 5627 (Gm5627).
Conclusions
The identified compound can serve as a potential therapeutic drug for the treatment of obesity and its related metabolic disorders. Furthermore, our newly clarified thermogenic genes play an important role in UCP1-dependent thermogenesis in adipocytes.
CONFLICT OF INTEREST STATEMENT
The authors declared no conflict of interest.
Supporting Information
Filename | Description |
---|---|
oby23948-sup-0001-supinfo.docxWord 2007 document , 693.9 KB | Data S1. Supporting Information |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
REFERENCES
- 1Bluher M. Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol. 2019; 15: 288-298.
- 2Ikeda K, Maretich P, Kajimura S. The common and distinct features of brown and beige adipocytes. Trends Endocrinol Metab. 2018; 29: 191-200.
- 3Chouchani ET, Kajimura S. Metabolic adaptation and maladaptation in adipose tissue. Nat Metab. 2019; 1: 189-200.
- 4Hasegawa Y. New perspectives on obesity-induced adipose tissue fibrosis and related clinical manifestations. Endocr J. 2022; 69: 739-748.
- 5Yamada T, Katagiri H, Ishigaki Y, et al. Signals from intra-abdominal fat modulate insulin and leptin sensitivity through different mechanisms: neuronal involvement in food-intake regulation. Cell Metab. 2006; 3: 223-229.
- 6Harms M, Seale P. Brown and beige fat: development, function and therapeutic potential. Nat Med. 2013; 19: 1252-1263.
- 7Virtanen KA, Lidell ME, Orava J, et al. Functional brown adipose tissue in healthy adults. N Engl J Med. 2009; 360: 1518-1525.
- 8Shinoda K, Luijten IH, Hasegawa Y, et al. Genetic and functional characterization of clonally derived adult human brown adipocytes. Nat Med. 2015; 21: 389-394.
- 9Yoneshiro T, Aita S, Matsushita M, et al. Recruited brown adipose tissue as an antiobesity agent in humans. J Clin Invest. 2013; 123: 3404-3408.
- 10Cohen P, Kajimura S. The cellular and functional complexity of thermogenic fat. Nat Rev Mol Cell Biol. 2021; 22: 393-409.
- 11Hasegawa Y, Ikeda K, Chen Y, et al. Repression of adipose tissue fibrosis through a PRDM16-GTF2IRD1 complex improves systemic glucose homeostasis. Cell Metab. 2018; 27: 180-194. e186.
- 12Ishigaki Y, Katagiri H, Yamada T, et al. Dissipating excess energy stored in the liver is a potential treatment strategy for diabetes associated with obesity. Diabetes. 2005; 54: 322-332.
- 13Tsukita S, Yamada T, Uno K, et al. Hepatic glucokinase modulates obesity predisposition by regulating BAT thermogenesis via neural signals. Cell Metab. 2012; 16: 825-832.
- 14Wei G, Sun H, Liu JL, Dong K, Liu J, Zhang M. Indirubin, a small molecular deriving from connectivity map (CMAP) screening, ameliorates obesity-induced metabolic dysfunction by enhancing brown adipose thermogenesis and white adipose browning. Nutr Metab. 2020; 17: 21.
10.1186/s12986-020-00440-4 Google Scholar
- 15Lu HY, Wang N, Li X, Huang Y, Wang J, Zhao QC. Identification of new potent human uncoupling protein 1 (UCP1) agonists using virtual screening and in vitro approaches. Mol Inform. 2019; 38:e1900030.
- 16Kawarasaki S, Matsuo K, Kuwata H, et al. Screening of flavor compounds using Ucp1-luciferase reporter beige adipocytes identified 5-methylquinoxaline as a novel UCP1-inducing compounds. Biosci Biotechnol Biochem. 2021; 86: 380-389.
- 17Tsui L. Adipocyte-based high throughput screening for anti-obesity drug discovery: current status and future perspectives. SLAS Discov. 2022; 27: 375-383.
- 18Galmozzi A, Sonne SB, Altshuler-Keylin S, et al. ThermoMouse: an in vivo model to identify modulators of UCP1 expression in brown adipose tissue. Cell Rep. 2014; 9: 1584-1593.
- 19Ohno H, Shinoda K, Ohyama K, Sharp LZ, Kajimura S. EHMT1 controls brown adipose cell fate and thermogenesis through the PRDM16 complex. Nature. 2013; 504: 163-167.
- 20Berridge MV, Herst PM, Tan AS. Tetrazolium dyes as tools in cell biology: new insights into their cellular reduction. Biotechnol Annu Rev. 2005; 11: 127-152.
- 21Xiao C, Goldgof M, Gavrilova O, Reitman ML. Anti-obesity and metabolic efficacy of the beta3-adrenergic agonist, CL316243, in mice at thermoneutrality compared to 22 degrees C. Obesity. 2015; 23: 1450-1459.
- 22Ahmed BA, Ong FJ, Barra NG, et al. Lower brown adipose tissue activity is associated with non-alcoholic fatty liver disease but not changes in the gut microbiota. Cell Rep Med. 2021; 2: 100397.
- 23Shinoda K, Ohyama K, Hasegawa Y, et al. Phosphoproteomics identifies CK2 as a negative regulator of beige adipocyte thermogenesis and energy expenditure. Cell Metab. 2015; 22: 997-1008.
- 24Zhang Y, Xie C, Wang H, et al. Irisin exerts dual effects on browning and adipogenesis of human white adipocytes. Am J Physiol Endocrinol Metab. 2016; 311: E530-E541.
- 25Lu J, Chatterjee M, Schmid H, Beck S, Gawaz M. CXCL14 as an emerging immune and inflammatory modulator. J Inflamm. 2016; 13: 1.
10.1186/s12950-015-0109-9 Google Scholar
- 26Wang Q, Doerschuk CM. The p38 mitogen-activated protein kinase mediates cytoskeletal remodeling in pulmonary microvascular endothelial cells upon intracellular adhesion molecule-1 ligation. J Immunol. 2001; 166: 6877-6884.
- 27Jurek B, Neumann ID. The oxytocin receptor: from intracellular signaling to behavior. Physiol Rev. 2018; 98: 1805-1908.
- 28Cuadrado A, Nebreda AR. Mechanisms and functions of p38 MAPK signalling. Biochem J. 2010; 429: 403-417.
- 29Matsushita Y, Hasegawa Y, Takebe N, et al. Serum C-X-C motif chemokine ligand 14 levels are associated with serum C-peptide and fatty liver index in type 2 diabetes mellitus patients. J Diabetes Investig. 2021; 12: 1042-1049.
- 30Cereijo R, Gavaldà-Navarro A, Cairó M, et al. CXCL14, a brown adipokine that mediates brown-fat-to-macrophage communication in thermogenic adaptation. Cell Metab. 2018; 28: 750-763. e756.
- 31Massoudi M, Evans E, Miller DS. Thermogenic drugs for the treatment of obesity: screening using obese rats and mice. Ann Nutr Metab. 1983; 27: 26-37.
- 32Colman E. Dinitrophenol and obesity: an early twentieth-century regulatory dilemma. Regul Toxicol Pharmacol. 2007; 48: 115-117.
- 33Goldgof M, Xiao C, Chanturiya T, Jou W, Gavrilova O, Reitman ML. The chemical uncoupler 2,4-dinitrophenol (DNP) protects against diet-induced obesity and improves energy homeostasis in mice at thermoneutrality. J Biol Chem. 2014; 289: 19341-19350.
- 34Medak KD, McKie GL, Shamshoum H, Seguin I, Wright DC. The glucose lowering effects of CL 316,243 dissipate with repeated use and are rescued bycilostamide. Physiol Rep. 2022; 10:e15187.
- 35Ikeda K, Yamada T. UCP1 dependent and independent thermogenesis in brown and beige adipocytes. Front Endocrinol. 2020; 11: 498.
- 36Gavalda-Navarro A, Villarroya J, Cereijo R, Giralt M, Villarroya F. The endocrine role of brown adipose tissue: an update on actors and actions. Rev Endocr Metab Disord. 2022; 23: 31-41.
- 37Greenhill C. Low brown adipose tissue activity linked to NAFLD. Nat Rev Endocrinol. 2021; 17: 707.
- 38Cao W, Daniel KW, Robidoux J, et al. p38 mitogen-activated protein kinase is the central regulator of cyclic AMP-dependent transcription of the brown fat uncoupling protein 1 gene. Mol Cell Biol. 2004; 24: 3057-3067.
- 39Leiva M, Matesanz N, Pulgarin-Alfaro M, Nikolic I, Sabio G. Uncovering the role of p38 family members in adipose tissue physiology. Front Endocrinol. 2020; 11: 572089.
- 40Bostrom P, Wu J, Jedrychowski MP, et al. A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature. 2012; 481: 463-468.
- 41Moreno-Navarrete JM, Ortega F, Serrano M, et al. Irisin is expressed and produced by human muscle and adipose tissue in association with obesity and insulin resistance. J Clin Endocrinol Metab. 2013; 98: E769-E778.
- 42Zhang Y, Li R, Meng Y, et al. Irisin stimulates browning of white adipocytes through mitogen-activated protein kinase p38 MAP kinase and ERK MAP kinase signaling. Diabetes. 2014; 63: 514-525.
- 43Zhao XY, Li S, Wang GX, Yu Q, Lin JD. A long noncoding RNA transcriptional regulatory circuit drives thermogenic adipocyte differentiation. Mol Cell. 2014; 55: 372-382.