1
|
Gong H, Qi Y, Wu X, Wu L, Liu W, Chen H, Qiu J, Wen H, Shen Z, Wang Z, Zhang M, Xu Z, Xu Z, Sun B, Li X, Zhao Q. UCP1-inspired mitochondrial uncouplers: Design, synthesis and thermogenic activity studies. Bioorg Chem 2025; 161:108466. [PMID: 40319812 DOI: 10.1016/j.bioorg.2025.108466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 05/07/2025]
Abstract
Given that uncoupling protein 1 (UCP1) serves as the principal thermogenic effector in adipose tissue, and motivated by recent breakthroughs in its structural characterization, we tried to obtain the precursor compounds with optimal docking mode, tryptophan compounds, by virtual screening. Following the initial structural refinement of these derivatives, we synthesized a series of tryptophan-based compounds aimed at enhancing their functional properties. Notably, the tryptophan-derived compound ZGL-18 was found to effectively induce lipid consumption without causing toxicity in brown adipocytes at a concentration of 100 μmol/L. In addition, administration of ZGL-18 stimulates brown adipocytes, trigger a reduction in mitochondrial membrane potential. In vivo experiments showed that ZGL-18 at a dose of 100 mg/kg significantly enhanced thermogenesis and cold tolerance in mice with preserving core and skin temperature. ZGL-18 was found to efficiently enhance respiration as well as energy expenditure in cold environments, in addition to passing, and was non-toxic at a dose of 1000 mg/kg. Furthermore, we preliminarily confirmed the binding mode of ZGL-18 to UCP1 by molecular docking and molecular dynamics simulation. These results suggest that ZGL-18 might be a promising candidate for drug development targeting UCP1.
Collapse
Affiliation(s)
- Hao Gong
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Yiming Qi
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Xinyi Wu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Limeng Wu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Wenjie Liu
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, People's Republic of China
| | - Huanhua Chen
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Jingsong Qiu
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya, Hainan 572025, People's Republic of China
| | - Han Wen
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Zixian Shen
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Zhiya Wang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Mingzuo Zhang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Zonghe Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Zihua Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Bohang Sun
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Xiang Li
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China.
| | - Qingchun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China.
| |
Collapse
|
2
|
Zhao K, Meng L, Wang X, Sui W, Zhang Y. Uncoupling protein 1-mediated protective effects of β3-adrenergic receptor agonist on kidney fibrosis via promoting adipose tissue browning in diabetic mice. Int J Biol Macromol 2025; 309:142977. [PMID: 40210064 DOI: 10.1016/j.ijbiomac.2025.142977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/16/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Diabetes mellitus (DM) is a global health concern. Diabetic kidney disease (DKD) is a prevalent severe complication of DM and therapy is urgently needed. Adipose tissue (AT) plays a crucial role in the energy mediation through glucolipid metabolism. Mirabegron is a specific β3-adrenergic receptor agonist, which can activate thermogenesis in adipocytes, improve energy consumption, and increase insulin sensitivity and glucose tolerance. Therefore, mirabegron may play a role in DKD pathogenesis. However, its effects and precise mechanisms remain unclear. METHODS A DKD mouse model based on type 2 DM (T2DM) was constructed and treated with mirabegron. Mice with AT surgically removed and mice with uncoupling protein 1 (Ucp1) knockout were used to confirm whether thermogenesis induced by mirabegron was the key process. RESULTS Mirabegron promoted AT browning in DKD mice. Mirabegron increased insulin sensitivity, promoted glucolipid metabolism, reduced inflammatory factor levels in kidney tissue, and improved renal function and fibrosis in DKD mice. Notably, all of these benefits disappeared in AT-removed DKD mice or in Ucp1 knockout DKD mice. CONCLUSIONS Mirabegron protects against kidney fibrosis in DM mice by activating AT thermogenesis via the UCP1 pathway. Thus, mirabegron may provide a promising potential option for DKD therapy.
Collapse
MESH Headings
- Animals
- Uncoupling Protein 1/metabolism
- Uncoupling Protein 1/genetics
- Adrenergic beta-3 Receptor Agonists/pharmacology
- Mice
- Fibrosis
- Thiazoles/pharmacology
- Acetanilides/pharmacology
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/drug effects
- Male
- Thermogenesis/drug effects
- Kidney/pathology
- Kidney/drug effects
- Kidney/metabolism
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/drug therapy
- Diabetic Nephropathies/pathology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/pathology
- Mice, Knockout
- Receptors, Adrenergic, beta-3/metabolism
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Kunsheng Zhao
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Linlin Meng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinlu Wang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenhai Sui
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Yun Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
3
|
Son S, Xu C, Jang J, Dinh M, Skorobogatko Y, Fu H, Valentine JM, An G, Ying W, Yu RT, Downes M, Evans RM, Saltiel AR. Sympathetic activation of white adipose tissue recruits neutrophils to limit energy expenditure. RESEARCH SQUARE 2025:rs.3.rs-6414640. [PMID: 40321773 PMCID: PMC12047989 DOI: 10.21203/rs.3.rs-6414640/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Adipose tissue maintains energy homeostasis by storing lipids during nutrient surplus and releasing them through lipolysis in times of energy demand. While lipolysis is essential for short term metabolic adaptation, prolonged metabolic stress requires adaptive changes that preserve energy reserves. Here, we report that β-adrenergic activation of adipocytes induces a transient and depot-specific infiltration of neutrophils into white adipose tissue (WAT), particularly in lipid-rich visceral WAT. Neutrophil recruitment requires the stimulation of both lipolysis and p38 MAPK activation in adipocytes. Recruited neutrophils locally secrete IL-1β, which suppresses lipolysis and limits excessive energy expenditure. Neutrophil depletion or blockade of IL-1β production increased lipolysis, leading to reduced WAT mass upon repeated β3-adrenergic stimulation. Together, these findings reveal an unexpected role of neutrophil-derived IL-1β in preserving lipid stores during metabolic stress, highlighting a physiological function of innate immune cells in maintaining energy homeostasis.
Collapse
Affiliation(s)
- Seunghwan Son
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Cindy Xu
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Janice Jang
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Maddox Dinh
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Yuliya Skorobogatko
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Haipeng Fu
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Joseph M. Valentine
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Garam An
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Ruth T. Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, San Diego, CA, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, San Diego, CA, USA
| | - Ronald M. Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, San Diego, CA, USA
| | - Alan R. Saltiel
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
4
|
Blok NB, Myronovych A, McMahon G, Bozadjieva-Kramer N, Seeley RJ. The evolution of steatosis and fibrosis in mice on a MASH-inducing diet and the effects of housing temperature. Am J Physiol Endocrinol Metab 2025; 328:E513-E523. [PMID: 39998384 DOI: 10.1152/ajpendo.00401.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/05/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Obesity induction in mice requires high-fat diet exposure. Although hepatic steatosis develops, progression to inflammation and fibrosis, as in humans, requires prolonged exposure and additional dietary factors. Immunosuppression at room temperature may slow this progression. We evaluated thermoneutrality's effect on metabolic dysfunction-associated steatohepatitis (MASH) development using a fibrosis-inducing MASH [Gubra-Amylin NASH (GAN)] diet. Mice were fed either a MASH or chow diet and housed at room temperature or thermoneutrality. MASH diet groups were euthanized monthly from 4 to 7 mo. Serum markers of hepatic function were analyzed, and liver histology assessed steatosis, inflammation, ballooning [nonalcoholic fatty liver disease activity score (NAS) score], and fibrosis via Picrosirius Red staining. MASH diet increased body weight, liver-to-body mass ratio, and hepatic fat, with no difference between housing conditions. Housing temperature had minimal effects on MASH. Serum markers and hepatic fibrosis were similar across groups. NAS score was lower at 4 mo in thermoneutral MASH mice but not by 7 mo. Thermoneutrality did not significantly impact MASH development. These findings, alongside existing literature, suggest thermoneutral housing does not consistently enhance MASH progression in GAN MASH-fed mice.NEW & NOTEWORTHY The development of MASH in mice-does housing temperature make a real difference?
Collapse
Affiliation(s)
- Neil B Blok
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Andriy Myronovych
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Garrett McMahon
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Nadejda Bozadjieva-Kramer
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
- Research Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, United States
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
5
|
Morris R, Bu K, Han W, Wood S, Hernandez Velez PM, Ward J, Crescitelli A, Martin M, Cheng F. The Association Between Statin Drugs and Rhabdomyolysis: An Analysis of FDA Adverse Event Reporting System (FAERS) Data and Transcriptomic Profiles. Genes (Basel) 2025; 16:248. [PMID: 40149400 PMCID: PMC11942242 DOI: 10.3390/genes16030248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/27/2025] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Rhabdomyolysis, a dangerous breakdown of skeletal muscle, has been reported as an adverse event in those prescribed a statin therapy for the treatment of hypercholesterolemia. Statin drugs are some of the most prescribed treatments for elevated cholesterol levels. The purpose of this comparative study was to determine the association between the statin drugs used and the risk of rhabdomyolysis using the FDA Adverse Event Reporting System (FAERS) and transcriptomic data. METHODS A disproportionality analysis was performed to compare the risk of rhabdomyolysis between the reference statin drug (simvastatin) and the treatment group, with patient age assessed as a possible confounder. In addition, association rule mining was utilized to both identify other adverse events that frequently presented with rhabdomyolysis and identify possible drug-drug interactions (DDIs). Finally, public transcriptomic data were explored to identify the possible genetic underpinnings highlighting these differences in rhabdomyolysis risk across statins. RESULTS Rhabdomyolysis is a commonly reported adverse event for patients treated with statins, particularly those prescribed simvastatin. Simvastatin was associated with a more than 2-fold increased likelihood of rhabdomyolysis compared to other statins. Men were twice as likely to report rhabdomyolysis than women regardless of statin treatment, with the highest risk observed for pravastatin (ROR = 2.30, p < 0.001) and atorvastatin (ROR = 2.03, p < 0.0001). Several possible DDIs were identified, including furosemide/Lasix, allopurinol clopidogrel/Plavix, and pantoprazole, which may elevate rhabdomyolysis risk through impaired muscle function and delayed statin metabolism. Finally, nine myopathic genes were identified as possible regulators of statin-induced rhabdomyolysis, including DYSF, DES, PLEC, CAPN3, SCN4A, TNNT1, SDHA, MYH7, and PYGM in primary human muscle cells. CONCLUSIONS Simvastatin was associated with the highest risk of rhabdomyolysis. The risk of rhabdomyolysis was more pronounced in men than women. Several possible DDIs were identified including furosemide/Lasix, allopurinol clopidogrel/Plavix, and pantoprazole.
Collapse
Affiliation(s)
- Robert Morris
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33613, USA; (R.M.); (S.W.); (P.M.H.V.); (J.W.); (A.C.); (M.M.)
| | - Kun Bu
- Department of Mathematics & Statistics, College of Art and Science, University of South Florida, Tampa, FL 33620, USA; (K.B.); (W.H.)
| | - Weiru Han
- Department of Mathematics & Statistics, College of Art and Science, University of South Florida, Tampa, FL 33620, USA; (K.B.); (W.H.)
| | - Savanah Wood
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33613, USA; (R.M.); (S.W.); (P.M.H.V.); (J.W.); (A.C.); (M.M.)
| | - Paola M. Hernandez Velez
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33613, USA; (R.M.); (S.W.); (P.M.H.V.); (J.W.); (A.C.); (M.M.)
| | - Jacob Ward
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33613, USA; (R.M.); (S.W.); (P.M.H.V.); (J.W.); (A.C.); (M.M.)
| | - Ariana Crescitelli
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33613, USA; (R.M.); (S.W.); (P.M.H.V.); (J.W.); (A.C.); (M.M.)
| | - Madison Martin
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33613, USA; (R.M.); (S.W.); (P.M.H.V.); (J.W.); (A.C.); (M.M.)
| | - Feng Cheng
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33613, USA; (R.M.); (S.W.); (P.M.H.V.); (J.W.); (A.C.); (M.M.)
| |
Collapse
|
6
|
Carpentier AC, Blondin DP. Is stimulation of browning of human adipose tissue a relevant therapeutic target? ANNALES D'ENDOCRINOLOGIE 2024; 85:184-189. [PMID: 38871497 DOI: 10.1016/j.ando.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Brown adipose tissue (BAT) and beige adipose tissues are important contributors to cold-induced whole body thermogenesis in rodents. The documentation in humans of cold- and ß-adrenergic receptor agonist-stimulated BAT glucose uptake using positron emission tomography (PET) and of a decrease of this response in individuals with cardiometabolic disorders led to the suggestion that BAT/beige adipose tissues could be relevant targets for prevention and treatment of these conditions. In this brief review, we will critically assess this question by first describing the basic rationale for this affirmation, second by examining the evidence in human studies, and third by discussing the possible means to activate the thermogenic response of these tissues in humans.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Sherbrooke, Québec, Canada.
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
7
|
Ruswandi YAR, Lesmana R, Rosdianto AM, Gunadi JW, Goenawan H, Zulhendri F. Understanding the Roles of Selenium on Thyroid Hormone-Induced Thermogenesis in Adipose Tissue. Biol Trace Elem Res 2024; 202:2419-2441. [PMID: 37758980 DOI: 10.1007/s12011-023-03854-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
Brown adipose tissue (BAT) and white adipose tissue (WAT) are known to regulate lipid metabolism. A lower amount of BAT compared to WAT, along with adipose tissue dysfunction, can result in obesity. Studies have shown that selenium supplementation protects against adipocyte dysfunction, decreases WAT triglycerides, and increases BAT triiodothyronine (T3). In this review, we discuss the relationship between selenium and lipid metabolism regulation through selenoprotein deiodinases and the role of deiodinases and thyroid hormones in the induction of adipose tissue thermogenesis. Upon 22 studies included in our review, we found that studies investigating the relationship between selenium and deiodinases demonstrated that selenium supplementation affects the iodothyronine deiodinase 2 (DIO2) protein and the expression of its associated gene, DIO2, proportionally. However, its effect on DIO1 is inconsistent while its effect on DIO3 activity is not detected. Studies have shown that the activity of deiodinases especially DIO2 protein and DIO2 gene expression is increased along with other browning markers upon white adipose tissue browning induction. Studies showed that thermogenesis is stimulated by the thyroid hormone T3 as its activity is correlated to the expression of other thermogenesis markers. A proposed mechanism of thermogenesis induction in selenium supplementation is by autophagy control. However, more studies are needed to establish the role of T3 and autophagy in adipose tissue thermogenesis, especially, since some studies have shown that thermogenesis can function even when T3 activity is lacking and studies related to autophagy in adipose tissue thermogenesis have contradictory results.
Collapse
Affiliation(s)
- Yasmin Anissa R Ruswandi
- Graduate School of Master Program in Anti-Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Kabupaten Sumedang, West Java, Indonesia
| | - Ronny Lesmana
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang, KM.21, Hegarmanah, Kec. Jatinangor, Kabupaten Sumedang, West Java, 45363, Indonesia.
| | - Aziiz Mardanarian Rosdianto
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang, KM.21, Hegarmanah, Kec. Jatinangor, Kabupaten Sumedang, West Java, 45363, Indonesia
- Veterinary Medicine Study Program, Faculty of Medicine, Universitas Padjadjaran, Kabupaten Sumedang, West Java, Indonesia
| | - Julia Windi Gunadi
- Department of Physiology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Hanna Goenawan
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang, KM.21, Hegarmanah, Kec. Jatinangor, Kabupaten Sumedang, West Java, 45363, Indonesia
| | - Felix Zulhendri
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Kabupaten Sumedang, West Java, Indonesia
- Kebun Efi, Kabanjahe, 22171, North Sumatra, Indonesia
| |
Collapse
|
8
|
Gong L, Zhao S, Chu X, Yang H, Li Y, Wei S, Li F, Zhang Y, Li S, Jiang P. Assessment of cold exposure-induced metabolic changes in mice using untargeted metabolomics. Front Mol Biosci 2023; 10:1228771. [PMID: 37719264 PMCID: PMC10500074 DOI: 10.3389/fmolb.2023.1228771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/18/2023] [Indexed: 09/19/2023] Open
Abstract
Background: Cold exposure (CE) can effectively modulate adipose tissue metabolism and improve metabolic health. Although previous metabolomics studies have primarily focused on analyzing one or two samples from serum, brown adipose tissue (BAT), white adipose tissue (WAT), and liver samples, there is a significant lack of simultaneous analysis of multiple tissues regarding the metabolic changes induced by CE in mice. Therefore, our study aims to investigate the metabolic profiles of the major tissues involved. Methods: A total of 14 male C57BL/6J mice were randomly assigned to two groups: the control group (n = 7) and the CE group (n = 7). Metabolite determination was carried out using gas chromatography-mass spectrometry (GC-MS), and multivariate analysis was employed to identify metabolites exhibiting differential expression between the two groups. Results: In our study, we identified 32 discriminant metabolites in BAT, 17 in WAT, 21 in serum, 7 in the liver, 16 in the spleen, and 26 in the kidney, respectively. Among these metabolites, amino acids, fatty acids, and nucleotides emerged as the most significantly altered compounds. These metabolites were found to be associated with 12 differential metabolic pathways closely related to amino acids, fatty acids, and energy metabolism. Conclusion: Our study may provide valuable insights into the metabolic effects induced by CE, and they have the potential to inspire novel approaches for treating metabolic diseases.
Collapse
Affiliation(s)
| | - Shiyuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, China
| | - Xue Chu
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
| | - Hui Yang
- Tengzhou Central People’s Hospital, Tengzhou, China
| | - Yanan Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Shanshan Wei
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Graduate Department, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, China
| | - Fengfeng Li
- Tengzhou Central People’s Hospital, Tengzhou, China
| | - Yazhou Zhang
- Tengzhou Central People’s Hospital, Tengzhou, China
| | - Shuhui Li
- Tengzhou Central People’s Hospital, Tengzhou, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, China
| |
Collapse
|
9
|
Cao Y, Langer R, Ferrara N. Targeting angiogenesis in oncology, ophthalmology and beyond. Nat Rev Drug Discov 2023; 22:476-495. [PMID: 37041221 DOI: 10.1038/s41573-023-00671-z] [Citation(s) in RCA: 131] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2023] [Indexed: 04/13/2023]
Abstract
Angiogenesis is an essential process in normal development and in adult physiology, but can be disrupted in numerous diseases. The concept of targeting angiogenesis for treating diseases was proposed more than 50 years ago, and the first two drugs targeting vascular endothelial growth factor (VEGF), bevacizumab and pegaptanib, were approved in 2004 for the treatment of cancer and neovascular ophthalmic diseases, respectively. Since then, nearly 20 years of clinical experience with anti-angiogenic drugs (AADs) have demonstrated the importance of this therapeutic modality for these disorders. However, there is a need to improve clinical outcomes by enhancing therapeutic efficacy, overcoming drug resistance, defining surrogate markers, combining with other drugs and developing the next generation of therapeutics. In this Review, we examine emerging new targets, the development of new drugs and challenging issues such as the mode of action of AADs and elucidating mechanisms underlying clinical benefits; we also discuss possible future directions of the field.
Collapse
Affiliation(s)
- Yihai Cao
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institute, Stockholm, Sweden.
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Napoleone Ferrara
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
10
|
Zhang S, Chen J, Li Q, Zeng W. Opioid growth factor receptor promotes adipose tissue thermogenesis via enhancing lipid oxidation. LIFE METABOLISM 2023; 2:load018. [PMID: 39872016 PMCID: PMC11749475 DOI: 10.1093/lifemeta/load018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/23/2023] [Accepted: 05/01/2023] [Indexed: 01/29/2025]
Abstract
The thermogenic brown and beige adipocytes consume fatty acids and generate heat to maintain core body temperature in the face of cold challenges. Since their validated presence in humans, the activation of thermogenic fat has been an attractive target for treating obesity and related metabolic diseases. Here, we reported that the opioid growth factor receptor (Ogfr) was highly expressed in adipocytes and promoted thermogenesis. The mice with genetic deletion of Ogfr in adipocytes displayed an impaired capacity to counter environmental cold challenges. Meanwhile, Ogfr ablation in adipocytes led to reduced fatty acid oxidation, enhanced lipid accumulation, impaired glucose tolerance, and exacerbated tissue inflammation under chronic high-fat diet (HFD)-fed conditions. At the cellular level, OGFr enhanced the production of mitochondrial trifunctional protein subunit α (MTPα) and also interacted with MTPα, thus promoting fatty acid oxidation. Together, our study demonstrated the important role of OGFr in fatty acid metabolism and adipose thermogenesis.
Collapse
Affiliation(s)
- Shan Zhang
- Institute for Immunology, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| | - Jianhui Chen
- Institute for Immunology, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| | - Qingqing Li
- Institute for Immunology, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| | - Wenwen Zeng
- Institute for Immunology, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| |
Collapse
|
11
|
Jing J, Peng Y, Fan W, Han S, Peng Q, Xue C, Qin X, Liu Y, Ding Z. Obesity-induced oxidative stress and mitochondrial dysfunction negatively affect sperm quality. FEBS Open Bio 2023; 13:763-778. [PMID: 36866962 PMCID: PMC10068321 DOI: 10.1002/2211-5463.13589] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/20/2023] [Accepted: 03/01/2023] [Indexed: 03/04/2023] Open
Abstract
Obesity is a systemic metabolic disease that can induce male infertility or subfertility through oxidative stress. The aim of this study was to determine how obesity impairs sperm mitochondrial structural integrity and function, and reduces sperm quality in both overweight/obese men and mice on a high-fat diet (HFD). Mice fed the HFD demonstrated higher body weight and increased abdominal fat content than those fed the control diet. Such effects accompanied the decline in antioxidant enzymes, such as glutathione peroxidase (GPX) and catalase and superoxide dismutase (SOD) in testicular and epidydimal tissues. Moreover, malondialdehyde (MDA) content significantly increased in sera. Mature sperm in HFD mice demonstrated higher oxidative stress, including increased mitochondrial reactive oxygen species (ROS) levels and decreased protein expression of GPX1, which may impair mitochondrial structural integrity and reduce mitochondrial membrane potential (MMP) and ATP production. Moreover, cyclic AMPK phosphorylation status increased, whereas sperm motility declined in the HFD mice. Clinical studies demonstrated that being overweight/obese reduced SOD enzyme activity in the seminal plasma and increased ROS in sperm, accompanied by lower MMP and low-quality sperm. Furthermore, ATP content in the sperm was negatively correlated with increases in the BMI of all clinical subjects. In conclusion, our results suggest that excessive fat intake had similar disruptive effects on sperm mitochondrial structure and function, as well as oxidative stress levels in humans and mice, which in turn induced lower sperm motility. This agreement strengthens the notion that fat-induced increases in ROS and impaired mitochondrial function contribute to male subfertility.
Collapse
Affiliation(s)
- Jia Jing
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Yuanhong Peng
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Weimin Fan
- Reproductive Medical Center of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Siyang Han
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Qihua Peng
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Chunran Xue
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Xinran Qin
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Yue Liu
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Zhide Ding
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
12
|
Fang R, Yan L, Liao Z. Abnormal lipid metabolism in cancer-associated cachexia and potential therapy strategy. Front Oncol 2023; 13:1123567. [PMID: 37205195 PMCID: PMC10185845 DOI: 10.3389/fonc.2023.1123567] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/17/2023] [Indexed: 05/21/2023] Open
Abstract
Cancer-associated cachexia (CAC) is a major characteristic of advanced cancer, associates with almost all types of cancer. Recent studies have found that lipopenia is an important feature of CAC, and it even occurs earlier than sarcopenia. Different types of adipose tissue are all important in the process of CAC. In CAC patients, the catabolism of white adipose tissue (WAT) is increased, leading to an increase in circulating free fatty acids (FFAs), resulting in " lipotoxic". At the same time, WAT also is induced by a variety of mechanisms, browning into brown adipose tissue (BAT). BAT is activated in CAC and greatly increases energy expenditure in patients. In addition, the production of lipid is reduced in CAC, and the cross-talk between adipose tissue and other systems, such as muscle tissue and immune system, also aggravates the progression of CAC. The treatment of CAC is still a vital clinical problem, and the abnormal lipid metabolism in CAC provides a new way for the treatment of CAC. In this article, we will review the mechanism of metabolic abnormalities of adipose tissue in CAC and its role in treatment.
Collapse
Affiliation(s)
- Ruoxin Fang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
| | - Ling Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, Hubei, China
- *Correspondence: Zhengkai Liao, ; Ling Yan,
| | - Zhengkai Liao
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
- *Correspondence: Zhengkai Liao, ; Ling Yan,
| |
Collapse
|
13
|
Cao Y. Blood vessels in fat tissues and vasculature-derived signals in controlling lipid metabolism and metabolic disease. Chin Med J (Engl) 2022; 135:2647-2652. [PMID: 36382988 PMCID: PMC9943976 DOI: 10.1097/cm9.0000000000002406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 65 Stockholm, Sweden
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, China
| |
Collapse
|
14
|
Patel A, Dobbins T, Kong X, Patel R, Carter G, Harding L, Sparks RP, Patel NA, Cooper DR. Induction of beige-like adipocyte markers and functions in 3T3-L1 cells by Clk1 and PKCβII inhibitory molecules. J Cell Mol Med 2022; 26:4183-4194. [PMID: 35801494 PMCID: PMC9344812 DOI: 10.1111/jcmm.17345] [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: 02/19/2021] [Revised: 03/18/2022] [Accepted: 03/25/2022] [Indexed: 11/29/2022] Open
Abstract
Excessive dietary intake of fat results in its storage in white adipose tissue (WAT). Energy expenditure through lipid oxidation occurs in brown adipose tissue (BAT). Certain WAT depots can undergo a change termed beiging where markers that BAT express are induced. Little is known about signalling pathways inducing beiging. Here, inhibition of a signalling pathway regulating alternative pre‐mRNA splicing is involved in adipocyte beiging. Clk1/2/4 kinases regulate splicing by phosphorylating factors that process pre‐mRNA. Clk1 inhibition by TG003 results in beige‐like adipocytes highly expressing PGC1α and UCP1. SiRNA for Clk1, 2 and 4, demonstrated that Clk1 depletion increased UCP1 and PGC1α expression, whereas Clk2/4 siRNA did not. TG003‐treated adipocytes contained fewer lipid droplets, are smaller, and contain more mitochondria, resulting in proton leak increases. Additionally, inhibition of PKCβII activity, a splice variant regulated by Clk1, increased beiging. PGC1α is a substrate for both Clk1 and PKCβII kinases, and we surmised that inhibition of PGC1α phosphorylation resulted in beiging of adipocytes. We show that TG003 binds Clk1 more than Clk2/4 through direct binding, and PGC1α binds to Clk1 at a site close to TG003. Furthermore, we show that TG003 is highly specific for Clk1 across hundreds of kinases in our activity screen. Hence, Clk1 inhibition becomes a target for induction of beige adipocytes.
Collapse
Affiliation(s)
- Achintya Patel
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Tradd Dobbins
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Xiaoyuan Kong
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Rehka Patel
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Gay Carter
- J.A. Haley Research Service, Tampa, Florida, USA
| | - Linette Harding
- Department of Chemistry, University of South Florida, Tampa, Florida, USA
| | - Robert P Sparks
- Department of Chemistry, University of South Florida, Tampa, Florida, USA
| | - Niketa A Patel
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida, USA.,J.A. Haley Research Service, Tampa, Florida, USA
| | - Denise R Cooper
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida, USA.,J.A. Haley Research Service, Tampa, Florida, USA
| |
Collapse
|
15
|
Brown-fat-mediated tumour suppression by cold-altered global metabolism. Nature 2022; 608:421-428. [PMID: 35922508 PMCID: PMC9365697 DOI: 10.1038/s41586-022-05030-3] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/28/2022] [Indexed: 02/05/2023]
Abstract
Glucose uptake is essential for cancer glycolysis and is involved in non-shivering thermogenesis of adipose tissues1-6. Most cancers use glycolysis to harness energy for their infinite growth, invasion and metastasis2,7,8. Activation of thermogenic metabolism in brown adipose tissue (BAT) by cold and drugs instigates blood glucose uptake in adipocytes4,5,9. However, the functional effects of the global metabolic changes associated with BAT activation on tumour growth are unclear. Here we show that exposure of tumour-bearing mice to cold conditions markedly inhibits the growth of various types of solid tumours, including clinically untreatable cancers such as pancreatic cancers. Mechanistically, cold-induced BAT activation substantially decreases blood glucose and impedes the glycolysis-based metabolism in cancer cells. The removal of BAT and feeding on a high-glucose diet under cold exposure restore tumour growth, and genetic deletion of Ucp1-the key mediator for BAT-thermogenesis-ablates the cold-triggered anticancer effect. In a pilot human study, mild cold exposure activates a substantial amount of BAT in both healthy humans and a patient with cancer with mitigated glucose uptake in the tumour tissue. These findings provide a previously undescribed concept and paradigm for cancer therapy that uses a simple and effective approach. We anticipate that cold exposure and activation of BAT through any other approach, such as drugs and devices either alone or in combination with other anticancer therapeutics, will provide a general approach for the effective treatment of various cancers.
Collapse
|
16
|
Li C, Davis X, Lahni P, Stuck J, Williamson L, Kaplan J. Obesity protects against sepsis-induced and norepinephrine-induced white adipose tissue browning. Am J Physiol Endocrinol Metab 2021; 321:E433-E442. [PMID: 34370596 PMCID: PMC8461795 DOI: 10.1152/ajpendo.00380.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 11/22/2022]
Abstract
Sepsis is a dysregulated systemic response to infection and can lead to organ damage and death. Obesity is a significant problem worldwide and affects outcomes from sepsis. Our laboratory demonstrated that white adipose tissue (WAT) undergoes browning during sepsis, a process whereby WAT adopts a brown adipose tissue phenotype. However, this browning process was not observed in obese mice during sepsis. White adipose tissue browning is detrimental in patients with burn injury and cancer. We hypothesize that norepinephrine (NE) induces WAT browning in nonobese mice but not in obese mice similarly to sepsis-induced WAT browning. Six-week-old C57BL/6 male mice were randomized to a high-fat diet or normal diet. After 6-7 wk of feeding, polymicrobial sepsis was induced by cecal ligation and puncture (CLP). Norepinephrine was administered intraperitoneally via osmotic minipumps for 18 h or 72 h (no CLP) at which time tissue and plasma were harvested. Controls were mice that underwent CLP (no NE) with 18-h harvest. A separate group of mice underwent pretreatment with NE or vehicle infusion for 72 h, CLP was performed, and at 18 h had tissue and plasma harvested. Sepsis resulted in significant weight loss in both nonobese and obese mice. NE treatment alone caused weight loss in obese mice. Septic nonobese mice had higher uncoupling protein-1 (UCP1) expression compared with control and obese septic mice. NE treatment increased UCP1 expression in nonobese, but not obese mice. NE-treated obese septic mice had lower lung myeloperoxidase (MPO) activity, alanine aminotransferase (ALT), aspartate aminotransferase (AST), TNFα, and IL-6 levels compared with NE-treated nonobese septic mice. Obesity protects mice from septic-induced and NE-induced WAT browning.NEW & NOTEWORTHY White adipose tissue browning is detrimental in patients with burn injury and cancer. WAT browning occurs in nonobese mice and can be induced by β receptor norepinephrine infusion, but obese mice are resistant to sepsis-induced and norepinephrine-induced WAT browning. We propose that the lack of WAT browning and unchanged inflammatory cytokine response may contribute to the protection of obese mice from sepsis.
Collapse
Affiliation(s)
- Cheryl Li
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Xenia Davis
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Patrick Lahni
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joanna Stuck
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lauren Williamson
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jennifer Kaplan
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
17
|
Seeley RJ, MacDougald OA. Mice as experimental models for human physiology: when several degrees in housing temperature matter. Nat Metab 2021; 3:443-445. [PMID: 33767444 PMCID: PMC8987294 DOI: 10.1038/s42255-021-00372-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Standfirst: Some “species differences” between mouse and human can be diminished simply by housing mice at warmer temperatures. Failure to strategically turn up the thermostat may undermine translation of findings in mice into insights on human metabolic diseases.
Collapse
Affiliation(s)
- Randy J Seeley
- Departments of Surgery, Internal Medicine and Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA.
| | - Ormond A MacDougald
- Departments of Molecular & Integrative Physiology, and Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
18
|
Han YH, Kee JY, Hong SH. Gomisin A Alleviates Obesity by Regulating the Phenotypic Switch between White and Brown Adipocytes. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1929-1948. [PMID: 34961413 DOI: 10.1142/s0192415x21500919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although gomisin A (GA) alleviates cancer and inflammation, its anti-obesity effect and the underlying mechanism have not yet been elucidated. Therefore, in this study, we aimed to elucidate the anti-obesity effects of GA by investigating the phenotypic changes involved in the browning and whitening of adipocytes. Here, obesity was induced to C57BL/6J mice using a high-fat diet (HFD). We administrated GA and checked weight changes for 12 weeks. We found that GA decreased the weight of weight gain, epididymal white adipose tissue (eWAT), and liver in the mice. In addition, the administration of GA elevated the levels of high-density lipoprotein (HDL)-cholesterol in the mice serum. Moreover, even after 12 weeks of treatment with GA, it did not cause any hepatic and renal toxicity. However, we found that GA induced the browning of eWAT and inhibited the whitening of brown adipose tissue. We further confirmed the anti-obesity mechanism of GA using 3T3-L1 cells, the human adipose mesenchymal stem cells (hAMSCs), and primary brown adipocytes (BAs) in vitroexperiments. We found that GA suppressed adipogenesis via the activation of AMP-activated protein kinase (AMPK). Furthermore, GA-induced browning by increasing the expression levels of uncoupling protein 1 (UCP1) in hAMSCs. The results of our study indicate that GA can inhibit weight gain by regulating the phenotypic changes involved in the browning and whitening of adipose tissues, which makes it a potential therapeutic agent for the treatment of obesity.
Collapse
Affiliation(s)
- Yo-Han Han
- Department of Oriental Pharmacy, College of Pharmacy Wonkwang-Oriental, Medicines Research Institute Wonkwang University, 344-2, Shinyong-dong, Iksan, KR, Iksan South Korea
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30602, USA
| | - Ji-Ye Kee
- Department of Oriental Pharmacy, College of Pharmacy Wonkwang-Oriental, Medicines Research Institute Wonkwang University, 344-2, Shinyong-dong, Iksan, KR, Iksan South Korea
| | - Seung-Heon Hong
- Department of Oriental Pharmacy, College of Pharmacy Wonkwang-Oriental, Medicines Research Institute Wonkwang University, 344-2, Shinyong-dong, Iksan, KR, Iksan South Korea
| |
Collapse
|
19
|
Abstract
Adipose, or fat, tissue (AT) was once considered an inert tissue that primarily existed to store lipids, and was not historically recognized as an important organ in the regulation and maintenance of health. With the rise of obesity and more rigorous research, AT is now recognized as a highly complex metabolic organ involved in a host of important physiological functions, including glucose homeostasis and a multitude of endocrine capabilities. AT dysfunction has been implicated in several disease states, most notably obesity, metabolic syndrome and type 2 diabetes. The study of AT has provided useful insight in developing strategies to combat these highly prevalent metabolic diseases. This review highlights the major functions of adipose tissue and the consequences that can occur when disruption of these functions leads to systemic metabolic dysfunction.
Collapse
Affiliation(s)
- Innocence Harvey
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Anik Boudreau
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Jacqueline M Stephens
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.,Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
20
|
AlZaim I, Hammoud SH, Al-Koussa H, Ghazi A, Eid AH, El-Yazbi AF. Adipose Tissue Immunomodulation: A Novel Therapeutic Approach in Cardiovascular and Metabolic Diseases. Front Cardiovasc Med 2020; 7:602088. [PMID: 33282920 PMCID: PMC7705180 DOI: 10.3389/fcvm.2020.602088] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is a critical regulator of systemic metabolism and bodily homeostasis as it secretes a myriad of adipokines, including inflammatory and anti-inflammatory cytokines. As the main storage pool of lipids, subcutaneous and visceral adipose tissues undergo marked hypertrophy and hyperplasia in response to nutritional excess leading to hypoxia, adipokine dysregulation, and subsequent low-grade inflammation that is characterized by increased infiltration and activation of innate and adaptive immune cells. The specific localization, physiology, susceptibility to inflammation and the heterogeneity of the inflammatory cell population of each adipose depot are unique and thus dictate the possible complications of adipose tissue chronic inflammation. Several lines of evidence link visceral and particularly perivascular, pericardial, and perirenal adipose tissue inflammation to the development of metabolic syndrome, insulin resistance, type 2 diabetes and cardiovascular diseases. In addition to the implication of the immune system in the regulation of adipose tissue function, adipose tissue immune components are pivotal in detrimental or otherwise favorable adipose tissue remodeling and thermogenesis. Adipose tissue resident and infiltrating immune cells undergo metabolic and morphological adaptation based on the systemic energy status and thus a better comprehension of the metabolic regulation of immune cells in adipose tissues is pivotal to address complications of chronic adipose tissue inflammation. In this review, we discuss the role of adipose innate and adaptive immune cells across various physiological and pathophysiological states that pertain to the development or progression of cardiovascular diseases associated with metabolic disorders. Understanding such mechanisms allows for the exploitation of the adipose tissue-immune system crosstalk, exploring how the adipose immune system might be targeted as a strategy to treat cardiovascular derangements associated with metabolic dysfunctions.
Collapse
Affiliation(s)
- Ibrahim AlZaim
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Safaa H. Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon
| | - Houssam Al-Koussa
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Alaa Ghazi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Basic Medical Sciences, College of Medicine, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
21
|
Ye C, Duan J, Zhang X, Yao L, Song Y, Wang G, Li Q, Wang B, Ai D, Wang C, Zhu Y. Cold-induced Yes-associated-protein expression through miR-429 mediates the browning of white adipose tissue. SCIENCE CHINA-LIFE SCIENCES 2020; 64:404-418. [PMID: 32804340 DOI: 10.1007/s11427-020-1779-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
Targeting the white-to-brown fat conversion is important for developing potential strategies to counteract metabolic diseases; yet the mechanisms are not fully understood. Yes-associated-protein (YAP), a transcription co-activator, was demonstrated to regulate adipose tissue functions; however, its effects on browning of subcutaneous white adipose tissue (sWAT) are unclear. We demonstrated that YAP was highly expressed in cold-induced beige fat. Mechanistically, YAP was found as a target gene of miR-429, which downregulated YAP expression in vivo and in vitro. In addition, miR-429 level was decreased in cold-induced beige fat. Additionally, pharmacological inhibition of the interaction between YAP and transcriptional enhanced associate domains by verteporfin dampened the browning of sWAT. Although adipose tissue-specific YAP overexpression increased energy expenditure with increased basal uncoupling protein 1 expression, it had no additional effects on the browning of sWAT in young mice. However, we found age-related impairment of sWAT browning along with decreased YAP expression. Under these circumstances, YAP overexpression significantly improved the impaired WAT browning in middle-aged mice. In conclusion, YAP as a regulator of sWAT browning, was upregulated by lowering miR-429 level in cold-induced beige fat. Targeting the miR-429-YAP pathway could be exploited for therapeutic strategies for age-related impairment of sWAT browning.
Collapse
Affiliation(s)
- Chenji Ye
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China
| | - Jinjie Duan
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China
| | - Xuejiao Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China
| | - Liu Yao
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China
| | - Yayue Song
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China
| | - Guangyan Wang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China
| | - Qi Li
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China
| | - Biqing Wang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China
| | - Ding Ai
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China
| | - Chunjiong Wang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China. .,Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China.
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
22
|
Vonhögen IG, el Azzouzi H, Olieslagers S, Vasilevich A, de Boer J, Tinahones FJ, da Costa Martins PA, de Windt LJ, Murri M. MiR-337-3p Promotes Adipocyte Browning by Inhibiting TWIST1. Cells 2020; 9:cells9041056. [PMID: 32340411 PMCID: PMC7226112 DOI: 10.3390/cells9041056] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 01/04/2023] Open
Abstract
The prevalence of metabolic syndrome (MetS) and obesity is an alarming health issue worldwide. Obesity is characterized by an excessive accumulation of white adipose tissue (WAT), and it is associated with diminished brown adipose tissue (BAT) activity. Twist1 acts as a negative feedback regulator of BAT metabolism. Therefore, targeting Twist1 could become a strategy for obesity and metabolic disease. Here, we have identified miR-337-3p as an upstream regulator of Twist1. Increased miR-337-3p expression paralleled decreased expression of TWIST1 in BAT compared to WAT. Overexpression of miR-337-3p in brown pre-adipocytes provoked a reduction in Twist1 expression that was accompanied by increased expression of brown/mitochondrial markers. Luciferase assays confirmed an interaction between the miR-337 seed sequence and Twist1 3′UTR. The inverse relationship between the expression of TWIST1 and miR-337 was finally validated in adipose tissue samples from non-MetS and MetS subjects that demonstrated a dysregulation of the miR-337-Twist1 molecular axis in MetS. The present study demonstrates that adipocyte miR-337-3p suppresses Twist1 repression and enhances the browning of adipocytes.
Collapse
Affiliation(s)
- Indira G.C. Vonhögen
- Department of Molecular Genetics, Faculty of Sciences and Engineering, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands; (I.G.C.V.); (H.e.A.); (S.O.); (P.A.d.C.M.)
| | - Hamid el Azzouzi
- Department of Molecular Genetics, Faculty of Sciences and Engineering, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands; (I.G.C.V.); (H.e.A.); (S.O.); (P.A.d.C.M.)
- Department of Molecular Genetics, Erasmus University MC, 3015 GD Rotterdam, The Netherlands
| | - Servé Olieslagers
- Department of Molecular Genetics, Faculty of Sciences and Engineering, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands; (I.G.C.V.); (H.e.A.); (S.O.); (P.A.d.C.M.)
| | - Aliaksei Vasilevich
- BioInterface Science Group, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands; (A.V.); (J.d.B.)
| | - Jan de Boer
- BioInterface Science Group, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands; (A.V.); (J.d.B.)
| | - Francisco J. Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (F.J.T.); (M.M.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERObn, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Faculty of Medicine, University of Malaga, 29010 Malaga, Spain
| | - Paula A. da Costa Martins
- Department of Molecular Genetics, Faculty of Sciences and Engineering, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands; (I.G.C.V.); (H.e.A.); (S.O.); (P.A.d.C.M.)
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Leon J. de Windt
- Department of Molecular Genetics, Faculty of Sciences and Engineering, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands; (I.G.C.V.); (H.e.A.); (S.O.); (P.A.d.C.M.)
- Correspondence:
| | - Mora Murri
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (F.J.T.); (M.M.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERObn, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
23
|
Sun X, Feng X, Wu X, Lu Y, Chen K, Ye Y. Fat Wasting Is Damaging: Role of Adipose Tissue in Cancer-Associated Cachexia. Front Cell Dev Biol 2020; 8:33. [PMID: 32117967 PMCID: PMC7028686 DOI: 10.3389/fcell.2020.00033] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/15/2020] [Indexed: 12/11/2022] Open
Abstract
Loss of body weight, especially loss of adipose tissue and skeletal muscle weight, characterizes cancer-associated cachexia (CAC). Clinically, therapeutic options for CAC are limited due to the complicated signaling between cancer and other organs. Recent research advances show that adipose tissues play a critical role during thermogenesis, glucose homeostasis, insulin sensitivity, and lipid metabolism. Understanding the adipocyte lipolysis, the formation of beige adipocytes, and the activation of brown adipocytes is vital for novel therapies for metabolic syndromes like CAC. The system-level crosstalk between adipose tissue and other organs involves adipocyte lipolysis, white adipose tissue browning, and secreted factors and metabolites. Novel CAC animal models and accumulating molecular signaling knowledge have provided mechanisms that may ultimately be translated into future therapeutic possibilities that benefit CAC patients. This mini review discusses the role of adipose tissue in CAC development, mechanism, and therapy.
Collapse
Affiliation(s)
- Xiaoting Sun
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaogang Feng
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Xiaojing Wu
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, China
| | - Yongtian Lu
- Department of ENT, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Kaihong Chen
- Department of Cardiology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, China
| | - Ying Ye
- Department of Oral Implantology, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
24
|
Wang N, Lu HY, Li X, Du YJ, Meng WH, Ding HW, Zhao QC. ZW290 Increases Cold Tolerance by Inducing Thermogenesis via the Upregulation of Uncoupling Protein 1 in Brown Adipose Tissue In Vitro and In Vivo. Lipids 2020; 54:265-276. [PMID: 31087415 DOI: 10.1002/lipd.12148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 03/10/2019] [Accepted: 03/26/2019] [Indexed: 11/06/2022]
Abstract
To provide molecular evidence on the thermogenic mechanism of primary brown adipocytes, western blot analysis was used to detect brown adipose tissue (BAT)-specific gene expressions. BAT protects the mammals from hypothermia injury with a large amount of mitochondria and high expression of uncoupling Protein 1 (UCP1), which is the vital protein to determine the heat production in BAT. In our previous study, the compound ZW290 (the structure shown in Fig. 1) was obtained by molecular docking with a UCP1 inducer. In the present study, ZW290 not only significantly upregulated the expression of UCP1 protein (p < 0.01) and its related signaling pathway in the primary brown adipocytes, but also remarkably decreased the mitochondrial membrane potential and the concentration of adenosine triphosphate (ATP) (p < 0.01). Kunming (KM) mice were kept under acute cold exposure (-20°C) to evaluate the preventive and protective effects of ZW290 on cold injury, and revealed its regulating mechanism in vitro. The rectal and body temperatures of ZW290-treated mice were significantly higher than those of the control (or model) group both at room temperature and at -20°C (p < 0.001). Hematoxylin-eosin (HE) staining and immunohistochemistry indicated that ZW290 notably decreased the size of lipid droplets in BAT and increased the content of mitochondria and the expression of UCP1 in BAT and white adipose tissue (WAT). Furthermore, the survival rate showed that ZW290 could prolong the overall survival of mice. Therefore, we obtained the conclusion that ZW290 might transform energy into heat by inhibiting ATP synthesis and increasing the expression of UCP1. Additionally, ZW290 may enhance cold tolerance by increasing heat production through increasing the content of mitochondria and the expression of UCP1 in BAT and WAT.
Collapse
Affiliation(s)
- Nan Wang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, Wenhua Road No. 83, Shenhe District, Shenyang 110840, China.,Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Wenhua Road No. 103, Shenhe District, Shenyang 110016, China
| | - Hong-Yuan Lu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Wenhua Road No. 103, Shenhe District, Shenyang 110016, China
| | - Xiang Li
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, Wenhua Road No. 83, Shenhe District, Shenyang 110840, China
| | - Ya-Jie Du
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Wenhua Road No. 103, Shenhe District, Shenyang 110016, China
| | - Wei-Hong Meng
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, Wenhua Road No. 83, Shenhe District, Shenyang 110840, China
| | - Huai-Wei Ding
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Wenhua Road No. 103, Shenhe District, Shenyang 110016, China
| | - Qing-Chun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, Wenhua Road No. 83, Shenhe District, Shenyang 110840, China.,Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Wenhua Road No. 103, Shenhe District, Shenyang 110016, China
| |
Collapse
|
25
|
Elsukova EI. Two-Level Organization of Thermogenesis in Adipose Tissue: a Morphofunctional Hypothesis. J EVOL BIOCHEM PHYS+ 2019. [DOI: 10.1134/s0022093019050065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
|
27
|
Yasmeen R, Shen Q, Lee A, Leung JH, Kowdley D, DiSilvestro DJ, Xu L, Yang K, Maiseyeu A, Bal NC, Periasamy M, Fadda P, Ziouzenkova O. Epiregulin induces leptin secretion and energy expenditure in high-fat diet-fed mice. J Endocrinol 2018; 239:377-388. [PMID: 30400011 PMCID: PMC6226053 DOI: 10.1530/joe-18-0289] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 09/19/2018] [Indexed: 01/30/2023]
Abstract
Adipokine leptin regulates neuroendocrine circuits that control energy expenditure, thermogenesis and weight loss. However, canonic regulators of leptin secretion, such as insulin and malonyl CoA, do not support these processes. We hypothesize that epiregulin (EREG), a growth factor that is secreted from fibroblasts under thermogenic and cachexia conditions, induces leptin secretion associated with energy dissipation. The effects of EREG on leptin secretion were studied ex vivo, in the intra-abdominal white adipose tissue (iAb WAT) explants, as well as in vivo, in WT mice with diet-induced obesity (DIO) and in ob/ob mice. These mice were pair fed a high-fat diet and treated with intraperitoneal injections of EREG. EREG increased leptin production and secretion in a dose-dependent manner in iAb fat explants via the EGFR/MAPK pathway. After 2 weeks, the plasma leptin concentration was increased by 215% in the EREG-treated group compared to the control DIO group. EREG-treated DIO mice had an increased metabolic rate and core temperature during the active dark cycle and displayed cold-induced thermogenesis. EREG treatment reduced iAb fat mass, the major site of leptin protein production and secretion, but did not reduce the mass of the other fat depots. In the iAb fat, expression of genes supporting mitochondrial oxidation and thermogenesis was increased in EREG-treated mice vs control DIO mice. All metabolic and gene regulation effects of EREG treatment were abolished in leptin-deficient ob/ob mice. Our data revealed a new role of EREG in induction of leptin secretion leading to the energy expenditure state. EREG could be a potential target protein to regulate hypo- and hyperleptinemia, underlying metabolic and immune diseases.
Collapse
Affiliation(s)
- Rumana Yasmeen
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Qiwen Shen
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Aejin Lee
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Jacob H. Leung
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Devan Kowdley
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - David J. DiSilvestro
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Lu Xu
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
- Department of Minimally Invasive Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Kefeng Yang
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
- Department of Nutrition, School of Medicine, Shanghai Jiao Tong University, Shanghai, China. 200025
| | - Andrei Maiseyeu
- Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Naresh C. Bal
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Muthu Periasamy
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Paolo Fadda
- Nucleic Acid Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Ouliana Ziouzenkova
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
- Correspondence Ouliana Ziouzenkova, PhD, 1787 Neil Avenue, 331A Campbell Hall; Columbus, OH 43210, ; Telephone: 001 614 292 5034; Fax: 001 614 292 8880
| |
Collapse
|
28
|
Balampanis K, Chasapi A, Kourea E, Tanoglidi A, Hatziagelaki E, Lambadiari V, Dimitriadis G, Lambrou GI, Kalfarentzos F, Melachrinou M, Sotiropoulou-Bonikou G. Inter-tissue expression patterns of the key metabolic biomarker PGC-1α in severely obese individuals: Implication in obesity-induced disease. Hellenic J Cardiol 2018; 60:282-293. [PMID: 30138744 DOI: 10.1016/j.hjc.2018.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/29/2018] [Accepted: 08/03/2018] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE PGC-1α is already known as a significant regulator of mitochondrial biogenesis, oxidative phosphorylation and fatty acid metabolism. Our study focuses on the role of PGC1α in morbid obesity, in five different tissues, collected from 50 severely obese patients during planned bariatric surgery. METHODS The investigated tissues included subcutaneous adipose tissue (SAT), visceral adipose tissue (VAT), skeletal muscle (SM), extramyocellular adipose tissue (EMAT) and liver. PGC1α expression was investigated with immunohistochemistry and evaluated with microscopy. RESULTS Our findings highlighted significant positive inter-tissue correlations regarding PGC-1α expression between several tissue pairs (VAT-SAT, VAT-SM, VAT-EMAT, SAT-SM, SAT-EMAT, SM-EMAT). Moreover, we found significant negative correlations between PGC1α expression in VAT with CD68 expression in skeletal muscle and EMAT, implying a possible protective role of PGC1α against obesity-induced inflammation. CONCLUSION Unmasking the inter-tissue communication networks regarding PGC-1α expression in morbid obesity, will give more insight into its significant role in obesity-induced diseases. PGC1α could potentially represent a future preventive and therapeutic target against obesity-induced disease, probably through enhancing mitochondrial biogenesis and metabolism.
Collapse
Affiliation(s)
- Konstantinos Balampanis
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece; Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - Athina Chasapi
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | - Eleni Kourea
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | - Anna Tanoglidi
- Department of Clinical Pathology, Akademiska University, Uppsala, Sweden.
| | - Erifili Hatziagelaki
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - Vaia Lambadiari
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - George Dimitriadis
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - George I Lambrou
- First Department of Pediatrics, Choremeio Research Laboratory, National and Kapodistrian University of Athens, Medical School, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece.
| | - Fotios Kalfarentzos
- Department of Surgery, Medical School, University of Patras, 26500 Patras, Greece.
| | - Maria Melachrinou
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | | |
Collapse
|
29
|
Yan K, Chen W, Zhu H, Lin G, Pan H, Li N, Wang L, Yang H, Liu M, Gong F. Ileal Transposition Surgery Decreases Fat Mass and Improves Glucose Metabolism in Diabetic GK Rats: Possible Involvement of FGF21. Front Physiol 2018; 9:191. [PMID: 29593555 PMCID: PMC5854974 DOI: 10.3389/fphys.2018.00191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 02/23/2018] [Indexed: 12/14/2022] Open
Abstract
Objective: Ileal transposition (IT) surgery has been reported to improve glucose and lipid metabolism, and fibroblast growth factor 21 (FGF21) is a powerful metabolic regulator. In the present study, we aimed to investigate the effects of IT surgery on metabolism and its possible relationship with the FGF21 signaling pathway in diabetic Goto-Kakizaki (GK) rats. Methods: Ten-week-old male GK rats were subjected to IT surgery with translocation of a 10 cm ileal segment to the proximal jejunum (IT group) or sham surgery without the ileum transposition (Sham-IT group). Rats in the no surgery group did not receive any surgical intervention. Six weeks later, body weight, fat mass, fasting blood glucose (FBG), and serum levels of FGF21 and leptin were measured. The expression of the FGF21 signaling pathway and white adipose tissue (WAT) browning-related genes in the WAT and liver were evaluated by real-time reverse transcription polymerase chain reaction (RT-qPCR) and western blot. Results: IT surgery significantly decreased the body weights and FBG levels and increased the insulin sensitivity of GK rats. The total WAT mass of the IT rats showed a 41.5% reduction compared with the Sham-IT rats, and serum levels of FGF21 and leptin of the IT rats decreased by 26.3 and 61.7%, respectively (all P < 0.05). The mRNA levels of fibroblast growth factor receptor 1 (FGFR1) and its co-receptor β klotho (KLB) in the perirenal WAT (pWAT) of the IT rats were 1.4- and 2.4-fold that of the Sham-IT rats, respectively, and the FGFR1 protein levels were 1.7-fold of the Sham-IT rats (all P < 0.05). In accordance with the pWAT, the protein levels of FGFR1 and KLB in the epididymal WAT (eWAT) of the IT rats notably increased to 3.0- and 3.9-fold of the Sham-IT rats (P < 0.05). Furthermore, uncoupling protein 1 (UCP1) protein levels in the eWAT and pWAT of the IT rats also increased to 2.2- and 2.3-fold of the Sham-IT rats (P < 0.05). However, the protein levels of FGFR1 and KLB in the subcutaneous WAT (sWAT) of the IT rats decreased by 34.4 and 72.1%, respectively, compared with the Sham-IT rats (P < 0.05). In addition, the protein levels of FGF21 and KLB in the livers of IT rats were 3.9- and 2.3-fold of the Sham-IT rats (all P < 0.05). Conclusions: IT surgery significantly decreased fat mass and improved glucose metabolism in diabetic GK rats. These beneficial roles of IT surgery were probably associated with its stimulatory action on the expression of FGFR1 and KLB in both the eWAT and the pWAT, thereby promoting UCP1 expression in these tissues.
Collapse
Affiliation(s)
- Kemin Yan
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Weijie Chen
- Department of Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huijuan Zhu
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guole Lin
- Department of Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hui Pan
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Naishi Li
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Linjie Wang
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hongbo Yang
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Meijuan Liu
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Fengying Gong
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
30
|
Abstract
The intrinsic cellular and metabolic properties of an adipocyte are shaped by the specific niche in which it resides. The diverse and discrete locations of major and minor rodent adipose depots are depicted in Part I. In Part II, the molecular and functional characteristics of four major types of adipocytes are described. Identified functions of relatively understudied but undoubtedly important depots are also highlighted.
Collapse
Affiliation(s)
- Devika P Bagchi
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Isabel Forss
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Susanne Mandrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Ormond A MacDougald
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|