DNA-mediated UCP1 overexpression in adipose tissue: A promising anti-obesity gene therapy

Ze-Wei Zhao , Longyun Hu , Bigui Song , Qian Wu , Jiejing Lin , Qingqing Liu , Siqi Liu , Jin Li , Molin Wang , Jin Li , Zhonghan Yang

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (10) : e70491

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (10) : e70491 DOI: 10.1002/ctm2.70491
RESEARCH ARTICLE

DNA-mediated UCP1 overexpression in adipose tissue: A promising anti-obesity gene therapy

Author information +
History +
PDF

Abstract

Background: Obesity has emerged as a global health challenge. Although GLP-1 receptor agonists are showing considerable promise in weight loss, their clinical utility is partly limited by gastrointestinal adverse reactions and non-fat weight loss side effects. UCP1-mediated adipose thermogenesis is a critical process for body temperature maintenance and weight management. However, the lack of effective and specific adipose thermogenesis therapies has restricted its clinical application. We aimed to explore the potential of inducing adipose-specific UCP1 overexpression via modified plasmids as an innovative therapeutic approach for obesity.

Methods: We replaced the cytomegalovirus (CMV) promoter in the plasmids with two types of adipose-specific promoters: mouse adiponectin (mADP) promoter and human adiponectin (hADP) promoter, to selectively overexpress UCP1 in adipocytes. The expression level of UCP1, weight loss, metabolic homeostasis and adipose thermogenesis effects were evaluated by immunohistochemistry, western blot, weight measurements, thermography, and comprehensive lab animal monitoring system.

Results: The experiments demonstrated that the mADP promoter-modified plasmids failed to drive UCP1 overexpression. In contrast, the hADP promoter-modified Ucp1 overexpression (hADP-Ucp1 OE) plasmids achieved robust adipose-specific UCP1 protein expression both in vitro and in vivo. In vitro experiments revealed that delivery of the hADP promoter-modified UCP1 overexpression (hADP-UCP1 OE) plasmids reduced lipid droplet size and enhanced energy consumption in human adipocytes. In obese mice, administration of the hADP-Ucp1 OE plasmids resulted in significant weight loss and improved metabolic homeostasis.

Conclusions: These findings highlight the therapeutic potential of hADP-UCP1 OE plasmids in obesity management.

Keywords

adipose tissue / gene therapy / obesity / thermogenesis / UCP1

Cite this article

Download citation ▾
Ze-Wei Zhao, Longyun Hu, Bigui Song, Qian Wu, Jiejing Lin, Qingqing Liu, Siqi Liu, Jin Li, Molin Wang, Jin Li, Zhonghan Yang. DNA-mediated UCP1 overexpression in adipose tissue: A promising anti-obesity gene therapy. Clinical and Translational Medicine, 2025, 15(10): e70491 DOI:10.1002/ctm2.70491

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

World Obesity Federation. World Obesity Atlas 2025. 2025. https://www.worldobesity.org/resources/resource-library/world-obesity-atlas-2025

[2]

Ng M, Fleming T, Robinson M, et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2014; 384(9945): 766-781.

[3]

Lingvay I, Cohen RV, Roux CWL, Sumithran P. Obesity in adults. Lancet. 2024; 404(10456): 972-987.

[4]

Fournier F, Diaz-Marin R, Pilon F, et al. Obesity triggers tumoral senescence and renders poorly immunogenic malignancies amenable to senolysis. Proc Natl Acad Sci U S A. 2023; 120(1): e2209973120.

[5]

Powell-Wiley TM, Poirier P, Burke LE, et al. Obesity and cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2021; 143(21): e984-e1010.

[6]

Cotillard A, Poitou C, Torcivia A, et al. Adipocyte size threshold matters: link with risk of type 2 diabetes and improved insulin resistance after gastric bypass. J Clin Endocrinol Metab. 2014; 99(8): E1466-70.

[7]

Sakers A, De Siqueira MK, Seale P, Villanueva CJ. Adipose-tissue plasticity in health and disease. Cell. 2022; 185(3): 419-446.

[8]

Bluher M. Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol. 2019; 15(5): 288-298.

[9]

Chu DT, Minh Nguyet NT, Dinh TC, et al. An update on physical health and economic consequences of overweight and obesity. Diabetes Metab Syndr. 2018; 12(6): 1095-1100.

[10]

Finkelstein EA, Trogdon JG, Cohen JW, Dietz W. Annual medical spending attributable to obesity: payer-and service-specific estimates. Health Aff (Millwood). 2009; 28(5): w822-31.

[11]

Carlsson LMS, Sjoholm K, Jacobson P, et al. Life expectancy after bariatric surgery in the Swedish obese subjects study. N Engl J Med. 2020; 383(16): 1535-1543.

[12]

Yanovski SZ, Yanovski JA. Approach to obesity treatment in primary care: a review. JAMA Intern Med. 2024; 184(7): 818-829.

[13]

Gilden AH, Catenacci VA, Taormina JM. Obesity. Ann Intern Med. 2024; 177(5): Itc65-itc80.

[14]

Wilding JPH, Batterham RL, Calanna S, et al. Once-weekly semaglutide in adults with overweight or obesity. N Engl J Med. 2021; 384(11): 989-1002.

[15]

Rubino D, Abrahamsson N, Davies M, et al. Effect of continued weekly subcutaneous semaglutide vs placebo on weight loss maintenance in adults with overweight or obesity: the STEP 4 randomized clinical trial. Jama. 2021; 325(14): 1414-1425.

[16]

Weiskirchen R, Lonardo A. How ‘miracle’ weight-loss semaglutide promises to change medicine but can we afford the expense?. Br J Pharmacol. 2025; 182(8): 1651-1670.

[17]

Wadden TA, Bailey TS, Billings LK, et al. Effect of subcutaneous semaglutide vs placebo as an adjunct to intensive behavioral therapy on body weight in adults with overweight or obesity: the STEP 3 randomized clinical trial. Jama. 2021; 325(14): 1403-1413.

[18]

Davies M, Færch L, Jeppesen OK, et al. Semaglutide 2·4 mg once a week in adults with overweight or obesity, and type 2 diabetes (STEP 2): a randomised, double-blind, double-dummy, placebo-controlled, phase 3 trial. Lancet. 2021; 397(10278): 971-984.

[19]

Tobaiqy M. A review of serious adverse events linked with GLP-1 agonists in type 2 diabetes mellitus and obesity treatment. Pharmacol Rep. 2024; 76(5): 981-990.

[20]

Karagiannis T, Malandris K, Avgerinos I, et al. Subcutaneously administered tirzepatide vs semaglutide for adults with type 2 diabetes: a systematic review and network meta-analysis of randomised controlled trials. Diabetologia. 2024; 67(7): 1206-1222.

[21]

Neeland IJ, Linge J, Birkenfeld AL. Changes in lean body mass with glucagon-like peptide-1-based therapies and mitigation strategies. Diabetes Obes Metab. 2024; 26(Suppl 4): 16-27.

[22]

Bikou A, Dermiki-Gkana F, Penteris M, Constantinides TK, Kontogiorgis C. A systematic review of the effect of semaglutide on lean mass: insights from clinical trials. Expert Opin Pharmacother. 2024; 25(5): 611-619.

[23]

Yoshino M, Kayser BD, Yoshino J, et al. Effects of diet versus gastric bypass on metabolic function in diabetes. N Engl J Med. 2020; 383(8): 721-732.

[24]

Kraljevic M, Süsstrunk J, Wölnerhanssen BK, et al. Long-term outcomes of laparoscopic Roux-en-Y gastric bypass vs laparoscopic sleeve gastrectomy for obesity: the SM-BOSS randomized clinical trial. JAMA Surg. 2025; 160(4): 369-377.

[25]

Maxim M, Soroceanu RP, Vlăsceanu VI, et al. Dietary habits, obesity, and bariatric surgery: a review of impact and interventions. Nutrients. 2025; 17(3): 474.

[26]

Goubar T, Goubar C, Fenton-Lee D, Stefanidis A, Macdonald PS, Rushworth RL. A population-based study of bariatric surgery trends in Australia: variations reflect continuing inequities in access to surgery. Obes Surg. 2025; 35(3): 1026-1035.

[27]

Agarwal N, Sharma K. Optimizing micro and macro nutrients: navigating weight loss success and health risks in bariatric surgery for obesity management. Endocrinol Metab Clin North Am. 2025; 54(1): 135-147.

[28]

Müller TD, Blüher M, Tschöp MH, DiMarchi RD. Anti-obesity drug discovery: advances and challenges. Nat Rev Drug Discov. 2022; 21(3): 201-223.

[29]

Legaard GE, Lyngbæk MPP, Almdal TP, et al. Effects of different doses of exercise and diet-induced weight loss on beta-cell function in type 2 diabetes (DOSE-EX): a randomized clinical trial. Nat Metab. 2023; 5(5): 880-895.

[30]

Liu QK. Mechanisms of action and therapeutic applications of GLP-1 and dual GIP/GLP-1 receptor agonists. Front Endocrinol (Lausanne). 2024; 15: 1431292.

[31]

Kim KS, Park JS, Hwang E, et al. GLP-1 increases preingestive satiation via hypothalamic circuits in mice and humans. Science. 2024; 385(6707): 438-446.

[32]

Quan Y, Lu F, Zhang Y. Use of brown adipose tissue transplantation and engineering as a thermogenic therapy in obesity and metabolic disease. Obes Rev. 2024; 25(3): e13677.

[33]

Zhao Z, Liu S, Qian B, et al. CMKLR1 senses chemerin/resolvin E1 to control adipose thermogenesis and modulate metabolic homeostasis. Fundam Res. 2024; 4(3): 575-588.

[34]

Zhao Z, Hu L, Song B, et al. Constitutively active receptor ADGRA3 signaling induces adipose thermogenesis. eLife. 2024; 13: RP100205.

[35]

Li L, Ma L, Zhao Z, et al. IL-25-induced shifts in macrophage polarization promote development of beige fat and improve metabolic homeostasis in mice. PLoS Biol. 2021; 19(8): e3001348.

[36]

Qin H, Zhong Y, Huang J, Miao Y, Du M, Huang K. TRIM56 promotes white adipose tissue browning to attenuate obesity by degrading TLE3. Adv Sci (Weinh). 2025; 12(13): e2414073.

[37]

Cheng L, Wang J, Dai H, et al. Brown and beige adipose tissue: a novel therapeutic strategy for obesity and type 2 diabetes mellitus. Adipocyte. 2021; 10(1): 48-65.

[38]

Ricquier D. UCP1, the mitochondrial uncoupling protein of brown adipocyte: a personal contribution and a historical perspective. Biochimie. 2017; 134: 3-8.

[39]

Feldmann HM, Golozoubova V, Cannon B, Nedergaard J. UCP1 ablation induces obesity and abolishes diet-induced thermogenesis in mice exempt from thermal stress by living at thermoneutrality. Cell Metab. 2009; 9(2): 203-209.

[40]

Shabalina IG, Petrovic N, de Jong JMA, Kalinovich AV, Cannon B, Nedergaard J. UCP1 in brite/beige adipose tissue mitochondria is functionally thermogenic. Cell Reports. 2013; 5(5): 1196-1203.

[41]

Rahbani JF, Bunk J, Lagarde D, et al. Parallel control of cold-triggered adipocyte thermogenesis by UCP1 and CKB. Cell Metab. 2024; 36(3): 526-540. e7.

[42]

Keipert S, Gaudry MJ, Kutschke M, et al. Two-stage evolution of mammalian adipose tissue thermogenesis. Science. 2024; 384(6700): 1111-1117.

[43]

Blondin DP, Nielsen S, Kuipers EN, et al. human brown adipocyte thermogenesis is driven by beta2-AR stimulation. Cell Metab. 2020; 32(2): 287-300. e7.

[44]

Muller TD, Bluher M, Tschop MH, DiMarchi RD. Anti-obesity drug discovery: advances and challenges. Nat Rev Drug Discov. 2021; 21(3): 201-223.

[45]

Grundlingh J, Dargan PI, El-Zanfaly M, Wood DM. 2,4-dinitrophenol (DNP): a weight loss agent with significant acute toxicity and risk of death. J Med Toxicol. 2011; 7(3): 205-212.

[46]

Gu Y, Cui S, Wang Q, et al. A randomized, double-blind, placebo-controlled phase II study of hepatocyte growth factor in the treatment of critical limb ischemia. Mol Ther. 2019; 27(12): 2158-2165.

[47]

Henry TD, Hirsch AT, Goldman J, et al. Safety of a non-viral plasmid-encoding dual isoforms of hepatocyte growth factor in critical limb ischemia patients: a phase I study. Gene Ther. 2011; 18(8): 788-794.

[48]

Powell RJ, Goodney P, Mendelsohn FO, Moen EK, Annex BH. Safety and efficacy of patient specific intramuscular injection of HGF plasmid gene therapy on limb perfusion and wound healing in patients with ischemic lower extremity ulceration: results of the HGF-0205 trial. J Vasc Surg. 2010; 52(6): 1525-1530.

[49]

Ma L, Zhao Z, Guo X, et al. Tanshinone IIA and its derivative activate thermogenesis in adipocytes and induce “beiging” of white adipose tissue. Mol Cell Endocrinol. 2022; 544: 111557.

[50]

Xue K, Wu D, Wang Y, et al. The mitochondrial calcium uniporter engages UCP1 to form a thermoporter that promotes thermogenesis. Cell Metab. 2022; 34(9): 1325-1341. e6.

[51]

Shamsi F, Xue R, Huang TL, et al. FGF6 and FGF9 regulate UCP1 expression independent of brown adipogenesis. Nat Commun. 2020; 11(1): 1421.

[52]

Schindelin J, Arganda-Carreras I, Frise E, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods. 2012; 9(7): 676-682.

[53]

Reinisch I, Ghosh A, Noé F, et al. Unveiling adipose populations linked to metabolic health in obesity. Cell Metab. 2025; 37(3): 640-655. e4.

[54]

Kajimura S, Spiegelman BM, Seale P. Brown and beige fat: physiological roles beyond heat generation. Cell Metab. 2015; 22(4): 546-559.

[55]

Chondronikola M, Volpi E, Borsheim E, et al. Brown adipose tissue improves whole-body glucose homeostasis and insulin sensitivity in humans. Diabetes. 2014; 63(12): 4089-4099.

[56]

Lidell ME, Betz MJ, Leinhard OD, et al. Evidence for two types of brown adipose tissue in humans. Nat Med. 2013; 19(5): 631-634.

[57]

Harms M, Seale P. Brown and beige fat: development, function and therapeutic potential. Nat Med. 2013; 19(10): 1252-1263.

[58]

Saito M, Okamatsu-Ogura Y, Matsushita M, et al. High incidence of metabolically active brown adipose tissue in healthy adult humans: effects of cold exposure and adiposity. Diabetes. 2009; 58(7): 1526-1531.

[59]

Finlin BS, Memetimin H, Zhu B, et al. The β3-adrenergic receptor agonist mirabegron improves glucose homeostasis in obese humans. J Clin Invest. 2020; 130(5): 2319-2331.

[60]

O'Mara AE, Johnson JW, Linderman JD, et al. Chronic mirabegron treatment increases human brown fat, HDL cholesterol, and insulin sensitivity. J Clin Invest. 2020; 130(5): 2209-2219.

[61]

Larsen TM, Toubro S, van Baak MA, et al. Effect of a 28-d treatment with L-796568, a novel beta(3)-adrenergic receptor agonist, on energy expenditure and body composition in obese men. Am J Clin Nutr. 2002; 76(4): 780-788.

[62]

Beasley HK, Wanjalla CN, Kirabo A. β(2)ARs: double edge sword in heart function. Trends Mol Med. 2023; 29(6): 422-424.

[63]

Johansen OS, Ma T, Hansen JB, et al. Lipolysis drives expression of the constitutively active receptor GPR3 to induce adipose thermogenesis. Cell. 2021; 184(13): 3502.

[64]

Chen K, Cheong LY, Gao Y, et al. Adipose-targeted triiodothyronine therapy counteracts obesity-related metabolic complications and atherosclerosis with negligible side effects. Nat Commun. 2022; 13(1): 7838.

[65]

Li X, Le Y, Zhang Z, Nian X, Liu B, Yang X. Viral vector-based gene therapy. Int J Mol Sci. 2023; 24(9): 7736.

[66]

Zhang Y, Wu ZY. Gene therapy for monogenic disorders: challenges, strategies, and perspectives. J Genet Genomics. 2024; 51(2): 133-143.

[67]

Blaszkiewicz M, Tao T, Mensah-Arhin K, et al. Gene therapy approaches for obesity-induced adipose neuropathy: device-targeted AAV-mediated neurotrophic factor delivery to adipocytes in subcutaneous adipose. Mol Ther. 2024; 32(5): 1407-1424.

[68]

Michurina S, Stafeev I, Boldyreva M, et al. Transplantation of adipose-tissue-engineered constructs with CRISPR-Mediated UCP1 activation. Int J Mol Sci. 2023; 24(4): 3844.

[69]

Wang CH, Lundh M, Fu A, et al. CRISPR-engineered human brown-like adipocytes prevent diet-induced obesity and ameliorate metabolic syndrome in mice. Sci Transl Med. 2020; 12(558): eaaz8664.

[70]

Luís MA, Goes MAD, Santos FM, Mesquita J, Tavares-Ratado P, Tomaz CT. Plasmid gene therapy for monogenic disorders: challenges and perspectives. Pharmaceutics. 2025; 17(1): 104.

[71]

Zhao Z, Yang Z. Reduction of serum lipids by liver-specific knockout of chemerin in DIO mice. Biochem Biophys Res Commun. 2025; 778: 152321.

[72]

Nguyen HP, An K, Ito Y, et al. Implantation of engineered adipocytes suppresses tumor progression in cancer models. Nat Biotechnol. 2025.

[73]

Jiang S, Li H, Zhang L, et al. Generic Diagramming Platform (GDP): a comprehensive database of high-quality biomedical graphics. Nucleic Acids Res. 2024; 53(D1): D1670-D1676.

[74]

Uhlén M, Fagerberg L, Hallström BM, et al. Tissue-based map of the human proteome. Science. 2015; 347(6220): 1260419.

[75]

Consortium TG, Aguet F, Anand S, et al. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science. 2020; 369(6509): 1318-1330.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

9

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/