101
|
Hayashi Y, Shimizu I, Yoshida Y, Ikegami R, Suda M, Katsuumi G, Fujiki S, Ozaki K, Abe M, Sakimura K, Okuda S, Hayano T, Nakamura K, Walsh K, Jespersen NZ, Nielsen S, Scheele C, Minamino T. Coagulation factors promote brown adipose tissue dysfunction and abnormal systemic metabolism in obesity. iScience 2022; 25:104547. [PMID: 35754738 PMCID: PMC9218513 DOI: 10.1016/j.isci.2022.104547] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/11/2022] [Accepted: 06/02/2022] [Indexed: 12/31/2022] Open
Abstract
Brown adipose tissue (BAT) has a role in maintaining systemic metabolic health in rodents and humans. Here, we show that metabolic stress induces BAT to produce coagulation factors, which then-together with molecules derived from the circulation-promote BAT dysfunction and systemic glucose intolerance. When mice were fed a high-fat diet (HFD), the levels of tissue factor, coagulation Factor VII (FVII), activated coagulation Factor X (FXa), and protease-activated receptor 1 (PAR1) expression increased significantly in BAT. Genetic or pharmacological suppression of coagulation factor-PAR1 signaling in BAT ameliorated its whitening and improved thermogenic response and systemic glucose intolerance in mice with dietary obesity. Conversely, the activation of coagulation factor-PAR1 signaling in BAT caused mitochondrial dysfunction in brown adipocytes and systemic glucose intolerance in mice fed normal chow. These results indicate that BAT produces endogenous coagulation factors that mediate pleiotropic effects via PAR1 signaling under metabolic stress.
Collapse
Affiliation(s)
- Yuka Hayashi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan
- Corresponding author
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan
- Department of Advanced Senotherapeutics, Juntendo University Graduate School of Medicine, Tokyo 113-8431, Japan
| | - Ryutaro Ikegami
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan
| | - Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan
| | - Shinya Fujiki
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Kazuyuki Ozaki
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuo-ku, Niigata 951-8585, Japan
- Department of Animal Model Development, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuo-ku, Niigata 951-8585, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuo-ku, Niigata 951-8585, Japan
- Department of Animal Model Development, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuo-ku, Niigata 951-8585, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Toshiya Hayano
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Shiga 525-8577 Japan
| | - Kazuhiro Nakamura
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kenneth Walsh
- Division of Cardiovascular Medicine, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Naja Zenius Jespersen
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, Copenhagen, Denmark
| | - Søren Nielsen
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, Copenhagen, Denmark
| | - Camilla Scheele
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
- Corresponding author
| |
Collapse
|
102
|
Zhang P, Wu S, He Y, Li X, Zhu Y, Lin X, Chen L, Zhao Y, Niu L, Zhang S, Li X, Zhu L, Shen L. LncRNA-Mediated Adipogenesis in Different Adipocytes. Int J Mol Sci 2022; 23:ijms23137488. [PMID: 35806493 PMCID: PMC9267348 DOI: 10.3390/ijms23137488] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Long-chain noncoding RNAs (lncRNAs) are RNAs that do not code for proteins, widely present in eukaryotes. They regulate gene expression at multiple levels through different mechanisms at epigenetic, transcription, translation, and the maturation of mRNA transcripts or regulation of the chromatin structure, and compete with microRNAs for binding to endogenous RNA. Adipose tissue is a large and endocrine-rich functional tissue in mammals. Excessive accumulation of white adipose tissue in mammals can cause metabolic diseases. However, unlike white fat, brown and beige fats release energy as heat. In recent years, many lncRNAs associated with adipogenesis have been reported. The molecular mechanisms of how lncRNAs regulate adipogenesis are continually investigated. In this review, we discuss the classification of lncRNAs according to their transcriptional location. lncRNAs that participate in the adipogenesis of white or brown fats are also discussed. The function of lncRNAs as decoy molecules and RNA double-stranded complexes, among other functions, is also discussed.
Collapse
Affiliation(s)
- Peiwen Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shuang Wu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuxu He
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinrong Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Zhu
- College of Life Science, China West Normal University, Nanchong 637009, China;
| | - Xutao Lin
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lei Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Ye Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lili Niu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shunhua Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xuewei Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Zhu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence: (L.Z.); (L.S.); Tel.: +86-28-8629-1133 (L.Z. & L.S.)
| | - Linyuan Shen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence: (L.Z.); (L.S.); Tel.: +86-28-8629-1133 (L.Z. & L.S.)
| |
Collapse
|
103
|
Huo C, Song Z, Yin J, Zhu Y, Miao X, Qian H, Wang J, Ye L, Zhou L. Effect of Acute Cold Exposure on Energy Metabolism and Activity of Brown Adipose Tissue in Humans: A Systematic Review and Meta-Analysis. Front Physiol 2022; 13:917084. [PMID: 35837014 PMCID: PMC9273773 DOI: 10.3389/fphys.2022.917084] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/13/2022] [Indexed: 11/29/2022] Open
Abstract
Background: The benefit of cold exposure for humans against obesity has brought the energy metabolism and activity of brown adipose tissue (BAT) induced by cold into focus. But the results are inconsistent. This review is aimed to systematically explore the effect of cold exposure on the activity of BAT and energy metabolism in humans. Methods: We searched relevant papers that were published from 1990 to 2021 and were cited in PubMed Central, Web of science, Embase and Cochrane Library databases to conduct this systematic review and meta-analysis. Energy metabolism, BAT volume, BAT activity and non-esterified fatty acids (NEFA) data reported in eligible researches were extracted. Meta-analysis was applied to combine the mean difference or standard mean difference with their 95% confidence intervals (95%CI). RevMan 5.3 software was used for meta-analysis and evaluating the risk of bias. Stata 16.0 was used for evaluating the publication bias. Results: Ten randomized controlled trials were included in meta-analysis. Compared with human exposed in room temperature at 24°C, the energy expenditure (EE) was increased after acute cold exposure at 16∼19°C (Z = 7.58, p < 0.05, mean different = 188.43kal/d, 95% CI = 139.73–237.13); BAT volume (Z = 2.62, p < 0.05; standard mean different = 0.41, 95% CI = 0.10–0.73); BAT activity (Z = 2.05, p = 0.04, standard mean difference = 1.61, 95% CI = 0.07–3.14) and the intake of BAT NEFA (Z = 2.85, p < 0.05; standard mean different = 0.53, 95% CI = 0.17–0.90) also increased. Conclusion: Acute cold exposure could improve the energy expenditure and BAT activity in adults, which is beneficial for human against obesity.
Collapse
Affiliation(s)
- Chuanyi Huo
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Zikai Song
- Department of Cardiology, the First Hospital of Jilin University, Changchun, China
| | - Jianli Yin
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Ying Zhu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Xiaohan Miao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Honghao Qian
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Jia Wang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Liting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
104
|
Takeda Y, Dai P. Chronic Fatty Acid Depletion Induces Uncoupling Protein 1 (UCP1) Expression to Coordinate Mitochondrial Inducible Proton Leak in a Human-Brown-Adipocyte Model. Cells 2022; 11:cells11132038. [PMID: 35805122 PMCID: PMC9265531 DOI: 10.3390/cells11132038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 11/17/2022] Open
Abstract
Thermogenic brown fat contributes to metabolic health in adult humans. Obese conditions are known to repress adipose-tissue browning and its activity. Herein, we found that chronic fatty acid (FA) depletion induced uncoupling protein 1 (UCP1) expression in the chemical-compound-induced brown adipocytes (ciBAs). The ciBAs, converted from human dermal fibroblasts under FA-free conditions, had low intracellular triglyceride levels and strongly activated UCP1 expression. Prolonged treatment with carnitine also reduced triglyceride accumulation and induced UCP1 expression. Transcriptome analysis revealed that the UCP1 induction was accompanied by the activation of lipid metabolic genes. The FA-depleted conditions repressed mitochondrial proton-leak activity and mitochondrial membrane potential (MMP), despite maintaining a high UCP1 expression. The evidence suggested that UCP1 expression was induced to compensate for the proton-leak activity under low MMP. Our study reports a regulatory mechanism underlying UCP1 expression and mitochondrial-energy status in human brown adipocytes under different nutritional conditions.
Collapse
Affiliation(s)
- Yukimasa Takeda
- Correspondence: (Y.T.); (P.D.); Tel.: +81-75-251-5444 (Y.T.); +81-75-251-5135 (P.D.)
| | - Ping Dai
- Correspondence: (Y.T.); (P.D.); Tel.: +81-75-251-5444 (Y.T.); +81-75-251-5135 (P.D.)
| |
Collapse
|
105
|
Tews D, Wabitsch M. Brown Adipose Tissue in Children and Its Metabolic Function. Horm Res Paediatr 2022; 95:104-111. [PMID: 34348306 DOI: 10.1159/000518353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/06/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND To regulate body temperature, mammals possess brown adipose tissue (BAT), which converts significant amounts of chemical energy into heat. Due to its remarkable energy demand, BAT is currently discussed as a target organ to treat obesity and obesity-related disorders. SUMMARY Although BAT is predominantly present in infants and its relative mass declines with age, new findings suggest that BAT has a relevant role in the regulation of energy homeostasis as well as in the regulation of the energy substrates glucose and lipids in older children, adolescents, and adults. In this overview, we will outline basic mechanisms of BAT thermogenesis and the recently described physiological relevance of BAT in metabolism in children and adolescents. KEY MESSAGE The connection of BAT activity with glucose metabolism and insulin sensitivity seems to be evident from recent studies, implicating BAT as an important influencing factor in the context of metabolic syndrome.
Collapse
Affiliation(s)
- Daniel Tews
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
106
|
Sugimoto S, Mena HA, Sansbury BE, Kobayashi S, Tsuji T, Wang CH, Yin X, Huang TL, Kusuyama J, Kodani SD, Darcy J, Profeta G, Pereira N, Tanzi RE, Zhang C, Serwold T, Kokkotou E, Goodyear LJ, Cypess AM, Leiria LO, Spite M, Tseng YH. Brown adipose tissue-derived MaR2 contributes to cold-induced resolution of inflammation. Nat Metab 2022; 4:775-790. [PMID: 35760872 PMCID: PMC9792164 DOI: 10.1038/s42255-022-00590-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 05/18/2022] [Indexed: 12/30/2022]
Abstract
Obesity induces chronic inflammation resulting in insulin resistance and metabolic disorders. Cold exposure can improve insulin sensitivity in humans and rodents, but the mechanisms have not been fully elucidated. Here, we find that cold resolves obesity-induced inflammation and insulin resistance and improves glucose tolerance in diet-induced obese mice. The beneficial effects of cold exposure on improving obesity-induced inflammation and insulin resistance depend on brown adipose tissue (BAT) and liver. Using targeted liquid chromatography with tandem mass spectrometry, we discovered that cold and β3-adrenergic stimulation promote BAT to produce maresin 2 (MaR2), a member of the specialized pro-resolving mediators of bioactive lipids that play a role in the resolution of inflammation. Notably, MaR2 reduces inflammation in obesity in part by targeting macrophages in the liver. Thus, BAT-derived MaR2 could contribute to the beneficial effects of BAT activation in resolving obesity-induced inflammation and may inform therapeutic approaches to combat obesity and its complications.
Collapse
Affiliation(s)
- Satoru Sugimoto
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Hebe Agustina Mena
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian E Sansbury
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shio Kobayashi
- Section of Immunobiology, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Tadataka Tsuji
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Chih-Hao Wang
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Xuanzhi Yin
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tian Lian Huang
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Joji Kusuyama
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Sean D Kodani
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Justin Darcy
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Gerson Profeta
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Nayara Pereira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Thomas Serwold
- Section of Immunobiology, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Efi Kokkotou
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Luiz Osório Leiria
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Matthew Spite
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
107
|
Pinto YO, Festuccia WTL, Magdalon J. The involvement of the adrenergic nervous system in activating human brown adipose tissue and browning. Hormones (Athens) 2022; 21:195-208. [PMID: 35247188 DOI: 10.1007/s42000-022-00361-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/24/2022] [Indexed: 11/04/2022]
Abstract
Obesity is a chronic condition of multifactorial etiology characterized by excessive body fat due to a calorie intake higher than energy expenditure. Given the intrinsic limitations of surgical interventions and the difficulties associated with lifestyle changes, pharmacological manipulation is currently one of the main therapies for metabolic diseases. Approaches aiming to promote energy expenditure through induction of thermogenesis have been explored and, in this context, brown adipose tissue (BAT) activation and browning have been shown to be promising strategies. Although such processes are physiologically stimulated by the sympathetic nervous system, not all situations that are known to increase adrenergic signaling promote a concomitant increase in BAT activation or browning in humans. Thus, a better understanding of factors involved in the thermogenesis attributed to these tissues is needed to enable the development of future therapies against obesity. Herein we carry out a critical review of original articles in humans under conditions previously known to trigger adrenergic responses-namely, cold, catecholamine-secreting tumor (pheochromocytoma and paraganglioma), burn injury, and adrenergic agonists-and discuss which of them are associated with increased BAT activation and browning. BAT is clearly stimulated in individuals exposed to cold or treated with high doses of the β3-adrenergic agonist mirabegron, whereas browning is certainly induced in patients after burn injury or with pheochromocytoma, as well as in individuals treated with β3-adrenergic agonist mirabegron for at least 10 weeks. Given the potential effect of increasing energy expenditure, adrenergic stimuli are promising strategies in the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Yolanda Oliveira Pinto
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, Sao Paulo, SP, Brazil
| | | | - Juliana Magdalon
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, Sao Paulo, SP, Brazil.
| |
Collapse
|
108
|
Ahmed BA, Varah N, Ong FJ, Blondin DP, Gunn E, Konyer NB, Singh NP, Noseworthy MD, Haman F, Carpentier AC, Punthakee Z, Steinberg GR, Morrison KM. Impaired Cold-Stimulated Supraclavicular Brown Adipose Tissue Activity in Young Boys With Obesity. Diabetes 2022; 71:1193-1204. [PMID: 35293989 DOI: 10.2337/db21-0799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022]
Abstract
Childhood obesity is a growing worldwide problem. In adults, lower cold-induced brown adipose tissue (BAT) activity is linked to obesity and metabolic dysfunction; this relationship remains uncertain in children. In this cross-sectional study, we compared cold-induced supraclavicular (SCV) BAT activity (percent change in proton density fat fraction [PDFF]) within the SCV region after 1 h of whole-body cold exposure (18°C), using MRI in 26 boys aged 8-10 years: 13 with normal BMI and 13 with overweight/obesity. Anthropometry, body composition, hepatic fat, visceral adipose tissue (VAT), and pre- and postcold PDFF of the subcutaneous adipose tissue (SAT) in the posterior neck region and the abdomen were measured. Boys with overweight/obesity had lower cold-induced percent decline in SCV PDFF compared with those with normal BMI (1.6 ± 0.8 vs. 4.7 ± 1.2%, P = 0.044). SCV PDFF declined significantly in boys with normal BMI (2.7 ± 0.7%, P = 0.003) but not in boys with overweight/obesity (1.1 ± 0.5%, P = 0.053). No cold-induced changes in the PDFF of either neck SAT (-0.89 ± 0.7%, P = 0.250, vs. 0.37 ± 0.3%, P = 0.230) or abdominal SAT (-0.39 ± 0.5%, P = 0.409, and 0.25 ± 0.2%, P = 0.139, for normal BMI and overweight/obesity groups, respectively) were seen. The cold-induced percent decline in SCV PDFF was inversely related to BMI (r = -0.39, P = 0.047), waist circumference (r = -0.48, P = 0.014), and VAT (r = -0.47, P = 0.014). Thus, in young boys, as in adults, BAT activity is lower in those with overweight/obesity, suggesting that restoring activity may be important for improving metabolic health.
Collapse
Affiliation(s)
- Basma A Ahmed
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Nina Varah
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Frank J Ong
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Elizabeth Gunn
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Norman B Konyer
- Imaging Research Centre, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Nina P Singh
- Department of Radiology, McMaster University, Hamilton, Ontario, Canada
| | - Michael D Noseworthy
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Imaging Research Centre, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
- Department of Radiology, McMaster University, Hamilton, Ontario, Canada
- Department of Electrical and Computer Engineering, McMaster University, Hamilton, Ontario, Canada
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Francois Haman
- Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Andre C Carpentier
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Zubin Punthakee
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Katherine M Morrison
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
109
|
Haman F, Souza SCS, Castellani JW, Dupuis MP, Friedl KE, Sullivan-Kwantes W, Kingma BRM. Human vulnerability and variability in the cold: Establishing individual risks for cold weather injuries. Temperature (Austin) 2022; 9:158-195. [PMID: 36106152 PMCID: PMC9467591 DOI: 10.1080/23328940.2022.2044740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 01/08/2023] Open
Abstract
Human tolerance to cold environments is extremely limited and responses between individuals is highly variable. Such physiological and morphological predispositions place them at high risk of developing cold weather injuries [CWI; including hypothermia and/or non-freezing (NFCI) and freezing cold injuries (FCI)]. The present manuscript highlights current knowledge on the vulnerability and variability of human cold responses and associated risks of developing CWI. This review 1) defines and categorizes cold stress and CWI, 2) presents cold defense mechanisms including biological adaptations, acute responses and acclimatization/acclimation and, 3) proposes mitigation strategies for CWI. This body of evidence clearly indicates that all humans are at risk of developing CWI without adequate knowledge and protective equipment. In addition, we show that while body mass plays a key role in mitigating risks of hypothermia between individuals and populations, NFCI and FCI depend mainly on changes in peripheral blood flow and associated decrease in skin temperature. Clearly, understanding the large interindividual variability in morphology, insulation, and metabolism is essential to reduce potential risks for CWI between and within populations.
Collapse
Affiliation(s)
- François Haman
- Faculty of Health Sciences, University of Ottawa, Ottawa,Ontario, Canada
| | - Sara C. S. Souza
- Faculty of Health Sciences, University of Ottawa, Ottawa,Ontario, Canada
| | - John W. Castellani
- Thermal and Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Maria-P. Dupuis
- Faculty of Health Sciences, University of Ottawa, Ottawa,Ontario, Canada
| | - Karl E. Friedl
- Thermal and Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Wendy Sullivan-Kwantes
- Biophysics and Biomedical Modeling Division, Defence Research Development Canada-Toronto, Defence Research and Development Canada, Ontario, Canada
| | - Boris R. M. Kingma
- Netherlands Organization for Applied Scientific Research, Department of Human Performance, Unit Defence, Safety and Security, Soesterberg, The Netherlands
| |
Collapse
|
110
|
Straat ME, Martinez-Tellez B, Sardjoe Mishre A, Verkleij MMA, Kemmeren M, Pelsma ICM, Alcantara JMA, Mendez-Gutierrez A, Kooijman S, Boon MR, Rensen PCN. Cold-Induced Thermogenesis Shows a Diurnal Variation That Unfolds Differently in Males and Females. J Clin Endocrinol Metab 2022; 107:1626-1635. [PMID: 35176767 PMCID: PMC9113803 DOI: 10.1210/clinem/dgac094] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Indexed: 11/21/2022]
Abstract
CONTEXT Cold exposure mobilizes lipids to feed thermogenic processes in organs, including brown adipose tissue (BAT). In rodents, BAT metabolic activity exhibits a diurnal rhythm, which is highest at the start of the wakeful period. OBJECTIVE We investigated whether cold-induced thermogenesis displays diurnal variation in humans and differs between the sexes. METHODS This randomized crossover study included 24 young and lean male (n = 12) and female (n = 12) participants who underwent 2.5-hour personalized cooling using water-perfused mattresses in the morning (7:45 am) and evening (7:45 pm), with 1 day in between. We measured energy expenditure (EE) and supraclavicular skin temperature in response to cold exposure. RESULTS In males, cold-induced EE was higher in the morning than in the evening (+54% ± 10% vs +30% ± 7%; P = 0.05) but did not differ between morning and evening in females (+37% ± 9% vs +30% ± 10%; P = 0.42). Only in males, supraclavicular skin temperature upon cold increased more in morning than evening (+0.2 ± 0.1 °C vs -0.2 ± 0.2 °C; P = 0.05). In males, circulating free fatty acid (FFA) levels were increased after morning cold exposure, but not evening (+90% ± 18% vs +9% ± 8%; P < 0.001). In females, circulating FFA (+94% ± 21% vs +20% ± 5%; P = 0.006), but also triglycerides (+42% ± 5% vs +29% ± 4%, P = 0.01) and cholesterol levels (+17% ± 2% vs 11% ± 2%; P = 0.05) were more increased after cold exposure in morning than in evening. CONCLUSION Cold-induced thermogenesis is higher in morning than evening in males; however, lipid metabolism is more modulated in the morning than the evening in females.
Collapse
Affiliation(s)
- Maaike E Straat
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Borja Martinez-Tellez
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Aashley Sardjoe Mishre
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Department of Radiology, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Magdalena M A Verkleij
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Mirjam Kemmeren
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Iris C M Pelsma
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Juan M A Alcantara
- PROFITH “PROmoting FITness and Health Through Physical Activity” Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | - Andrea Mendez-Gutierrez
- Department of Biochemistry and Molecular Biology II, “José Mataix Verdú” Institute of Nutrition and Food Technology, Center of Biomedical Research, University of Granada, Granada, Spain
- Biohealth Research Institute in Granada (ibs.GRANADA), Granada, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Mariëtte R Boon
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| |
Collapse
|
111
|
Li A, Liao W, Xie J, Song L, Zhang X. Plasma Proteins as Occupational Hazard Risk Monitors for Populations Working in Harsh Environments: A Mendelian Randomization Study. Front Public Health 2022; 10:852572. [PMID: 35602164 PMCID: PMC9120921 DOI: 10.3389/fpubh.2022.852572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/30/2022] [Indexed: 11/25/2022] Open
Abstract
Harsh work environments can include very cold, hot, dusty, and noisy workplaces, as well as exposure in the workplace with chemicals and other fumes, cigarette smoke, and diesel exhaust. Although working in these harsh environments can have a negative effect on health, there are no effective biomarkers for monitoring health conditions until workers develop disease symptoms. Plasma protein concentrations, which reflect metabolism and immune status, have great potential as biomarkers for various health conditions. Using a Mendelian-randomization (MR) design, this study analyzed the effects of these harsh environments on plasma proteins to identify proteins that can be used as biomarkers of health status. Preliminary analysis using inverse variance weighted (IVW) method with a p-value cutoff of 0.05 showed that workplace environments could affect the concentrations of hundreds of plasma proteins. After filtering for sensitivity via MR-Egger, and Weighted Median MR approaches, 28 plasma proteins altered by workplace environments were identified. Further MR analysis showed that 20 of these plasma proteins, including UNC5D, IGFBP1, SCG3, ST3GAL6, and ST3GAL2 are affected by noisy workplace environments; TFF1, RBM39, ACYP2, STAT3, GRB2, CXCL1, EIF1AD, CSNK1G2, and CRKL that are affected by chemical fumes; ADCYAP1, NRSN1, TMEM132A, and CA10 that are affected by passive smoking; LILRB2, and TENM4 that are affected by diesel exhaust, are associated with the risk of at least one disease. These proteins have the potential to serve as biomarkers to monitor the occupational hazards risk of workers working in corresponding environments. These findings also provide clues to study the biological mechanisms of occupational hazards.
Collapse
Affiliation(s)
- Ang Li
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenjing Liao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Junyang Xie
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijuan Song
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaowen Zhang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangzhou, China
- *Correspondence: Xiaowen Zhang
| |
Collapse
|
112
|
Frankl JA, An Y, Sherwood A, Hao G, Huang FY, Thapa P, Clegg DJ, Sun X, Scherer PE, Öz OK. Comparison of BMIPP-SPECT/CT to 18FDG-PET/CT for Imaging Brown or Browning Fat in a Preclinical Model. Int J Mol Sci 2022; 23:4880. [PMID: 35563272 PMCID: PMC9101718 DOI: 10.3390/ijms23094880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 11/17/2022] Open
Abstract
Obesity is a leading cause of preventable death and morbidity. To elucidate the mechanisms connecting metabolically active brown adipose tissue (BAT) and metabolic health may provide insights into methods of treatment for obesity-related conditions. 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18FDG-PET/CT) is traditionally used to image human BAT activity. However, the primary energy source of BAT is derived from intracellular fatty acids and not glucose. Beta-methyl-p-iodophenylpentadecanoic acid (BMIPP) is a fatty acid analogue amenable to in vivo imaging by single photon emission computed tomography/CT (SPECT/CT) when radiolabeled with iodine isotopes. In this study, we compare the use of 18FDG-PET/CT and 125I-BMIPP-SPECT/CT for fat imaging to ascertain whether BMIPP is a more robust candidate for the non-invasive evaluation of metabolically active adipose depots. Interscapular BAT, inguinal white adipose tissue (iWAT), and gonadal white adipose tissue (gWAT) uptake of 18FDG and 125I-BMIPP was quantified in mice following treatment with the BAT-stimulating drug CL-316,243 or saline vehicle control. After CL-316,243 treatment, uptake of both radiotracers increased in BAT and iWAT. The standard uptake value (SUVmean) for 18FDG and 125I-BMIPP significantly correlated in these depots, although uptake of 125I-BMIPP in BAT and iWAT more closely mimicked the fold-change in metabolic rate as measured by an extracellular flux analyzer. Herein, we find that imaging BAT with the radioiodinated fatty acid analogue BMIPP yields more physiologically relevant data than 18FDG-PET/CT, and its conventional use may be a pivotal tool for evaluating BAT in both mice and humans.
Collapse
Affiliation(s)
- Joseph A. Frankl
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA; (J.A.F.); (A.S.); (G.H.); (P.T.); (X.S.)
| | - Yu An
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Amber Sherwood
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA; (J.A.F.); (A.S.); (G.H.); (P.T.); (X.S.)
| | - Guiyang Hao
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA; (J.A.F.); (A.S.); (G.H.); (P.T.); (X.S.)
| | - Feng-Yun Huang
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, No. 666, Buzih Road, Beitun District, Taichung City 406053, Taiwan;
| | - Pawan Thapa
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA; (J.A.F.); (A.S.); (G.H.); (P.T.); (X.S.)
| | - Deborah J. Clegg
- Department of Internal Medicine, Texas Tech Health Sciences Center, 5001 El Paso Dr, El Paso, TX 79905, USA;
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA; (J.A.F.); (A.S.); (G.H.); (P.T.); (X.S.)
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, Southwestern Medical Center, University of Texas, 5323 Harry Hines Blvd, Dallas, TX 75390, USA;
| | - Orhan K. Öz
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA; (J.A.F.); (A.S.); (G.H.); (P.T.); (X.S.)
| |
Collapse
|
113
|
Crandall J, Fraum TJ, Wahl RL. Brown adipose tissue: a protective mechanism in "pre-prediabetes"? J Nucl Med 2022; 63:1433-1440. [PMID: 35393347 DOI: 10.2967/jnumed.121.263357] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 03/21/2022] [Indexed: 11/16/2022] Open
Abstract
Brown adipose tissue (BAT) is present in a significant number of adult humans and has been postulated to exert beneficial metabolic effects. Lean, non-diabetic patients undergoing clinical positron emission tomography (PET)/computed tomography (CT) imaging are more likely to exhibit incidental BAT activation. The aim of this study was to assess metabolic changes associated with the cold-activation of BAT and to compare baseline blood metabolites in participants with varying amounts of active BAT. Methods: Serum blood samples were collected from healthy adult volunteers (body mass index 18.0-25.0 and age≤35 years) before and after 2 h cold exposure. 18F-flurodeoxyglucose (FDG) PET/CT imaging was performed immediately following cold exposure. Activated BAT was segmented and fasting glucose, insulin, lipid, and other blood metabolite levels were correlated with volume and intensity of active BAT. Using a median cutoff, subjects were classified as BATHIGH or BATLOW. Results: A higher volume of activated BAT was associated with significantly higher pre-cooling glucose and insulin levels (P<0.001 for each). Pre-cooling thyroid stimulating hormone (TSH) and triglyceride levels were significantly higher in the BATHIGH than in the BATLOW group (P = 0.002 and P<0.001, respectively). Triglyceride levels tended to increase over the cooling period in both BAT groups, but increased significantly more in the BATHIGH group (15.7±13.2 md/dl; P<0.001) than in the BATLOW group (4.5±12.2 mg/dl; P = 0.061). Conclusion: These findings may indicate that BAT is recruited to counteract incipient "pre-prediabetic" states, potentially serving as a first-line protective mechanism against very early metabolic or hormonal variations.
Collapse
|
114
|
Sanders KJC, Wierts R, van Marken Lichtenbelt WD, de Vos-Geelen J, Plasqui G, Kelders MCJM, Schrauwen-Hinderling VB, Bucerius J, Dingemans AMC, Mottaghy FM, Schols AMWJ. Brown adipose tissue activation is not related to hypermetabolism in emphysematous chronic obstructive pulmonary disease patients. J Cachexia Sarcopenia Muscle 2022; 13:1329-1338. [PMID: 35166050 PMCID: PMC8978002 DOI: 10.1002/jcsm.12881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 09/27/2021] [Accepted: 11/01/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Brown adipose tissue (BAT) has been primarily researched as a potential target for mitigating obesity. However, the physiological significance of BAT in relation to cachexia remains poorly understood. The objective of this study was to investigate the putative contribution of BAT on different components of energy metabolism in emphysematous chronic obstructive pulmonary disease (COPD) patients. METHODS Twenty COPD patients (mean ± SD age 62 ± 6, 50% female, median [range] BMI 22.4 [15.1-32.5] kg/m2 and 85% low FFMI) were studied. Basal metabolic rate (BMR) was assessed by ventilated hood, total daily energy expenditure (TDEE) by doubly labelled water and physical activity by triaxial accelerometry. BMR was adjusted for fat-free mass (FFM) as assessed by deuterium dilution. Analysis of BAT and WAT was conducted in a subset of ten patients and six age-matched, gender-matched and BMI-matched healthy controls. BAT glucose uptake was assessed by means of cold-stimulated integrated [18F]FDG positron-emission tomography and magnetic resonance imaging. WAT was collected from subcutaneous abdominal biopsies to analyse metabolic and inflammatory gene expression levels. Lung function was assessed by spirometry and body plethysmography and systemic inflammation by high sensitivity C-reactive protein. RESULTS Mean TDEE was 2209 ± 394 kcal/day, and mean BMR was 1449 ± 214 kcal/day corresponding to 120% of predicted. FFM-adjusted BMR did not correlate with lung function or C-reactive protein. Upon cooling, energy expenditure increased, resulting in a non-shivering thermogenesis of (median [range]) 20.1% [3.3-41.3] in patients and controls. Mean BAT glucose uptake was comparable between COPD and controls (1.5 [0.1-6.2] vs. 1.1 [0.7-3.9]). In addition, no correlation was found between BMR adjusted for FFM and BAT activity or between cold-induced non-shivering energy expenditure and BAT activity. Gene expression levels of the brown adipocyte or beige markers were also comparable between the groups. No (serious) adverse events were reported. CONCLUSIONS Although COPD patients were hypermetabolic at rest, no correlation was found between BMR or TDEE and BAT activity. Furthermore, both BAT activity and gene expression levels of the brown adipocyte or beige markers were comparable between COPD patients and controls.
Collapse
Affiliation(s)
- Karin J C Sanders
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Roel Wierts
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Wouter D van Marken Lichtenbelt
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Judith de Vos-Geelen
- Department of Internal Medicine, Division of Medical Oncology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Guy Plasqui
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marco C J M Kelders
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Vera B Schrauwen-Hinderling
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Radiology and Nuclear Medicine, NUTRIM School for Nutrition and Translational Research in Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jan Bucerius
- Department of Radiology and Nuclear Medicine and CARIM School for Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Nuclear Medicine, University Medicine Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | | | - Felix M Mottaghy
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Nuclear Medicine and CIO ABCD, University Hospital RWTH Aachen University, Aachen, Germany
| | - Annemie M W J Schols
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
115
|
van Eenige R, In Het Panhuis W, Schönke M, Jouffe C, Devilee TH, Siebeler R, Streefland TCM, Sips HCM, Pronk ACM, Vorderman RHP, Mei H, van Klinken JB, van Weeghel M, Uhlenhaut NH, Kersten S, Rensen PCN, Kooijman S. Angiopoietin-like 4 governs diurnal lipoprotein lipase activity in brown adipose tissue. Mol Metab 2022; 60:101497. [PMID: 35413480 PMCID: PMC9048098 DOI: 10.1016/j.molmet.2022.101497] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/29/2022] Open
Abstract
Objective Brown adipose tissue (BAT) burns fatty acids (FAs) to produce heat, and shows diurnal oscillation in glucose and triglyceride (TG)-derived FA-uptake, peaking around wakening. Here we aimed to gain insight in the diurnal regulation of metabolic BAT activity. Methods RNA-sequencing, chromatin immunoprecipitation (ChIP)-sequencing, and lipidomics analyses were performed on BAT samples of wild type C57BL/6J mice collected at 3-hour intervals throughout the day. Knockout and overexpression models were used to study causal relationships in diurnal lipid handling by BAT. Results We identified pronounced enrichment of oscillating genes involved in extracellular lipolysis in BAT, accompanied by oscillations of FA and monoacylglycerol content. This coincided with peak lipoprotein lipase (Lpl) expression, and was predicted to be driven by peroxisome proliferator-activated receptor gamma (PPARγ) activity. ChIP-sequencing for PPARγ confirmed oscillation in binding of PPARγ to Lpl. Of the known LPL-modulators, angiopoietin-like 4 (Angptl4) showed the largest diurnal amplitude opposite to Lpl, and both Angptl4 knockout and overexpression attenuated oscillations of LPL activity and TG-derived FA-uptake by BAT. Conclusions Our findings highlight involvement of PPARγ and a crucial role of ANGPTL4 in mediating the diurnal oscillation of TG-derived FA-uptake by BAT, and imply that time of day is essential when targeting LPL activity in BAT to improve metabolic health. The transcriptome and lipidome of brown fat show clusters with distinct circadian phases. The peak in metabolic brown fat activity is defined by activation of lipolytic processes. PPARγ shows oscillating binding to lipolytic genes and may drive diurnal brown fat activity. Genetic modulation of the lipoprotein lipase inhibitor Angptl4 flattens rhythmic activity in brown fat. Time of day should be considered when studying the metabolic benefits of targeting brown fat.
Collapse
Affiliation(s)
- Robin van Eenige
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Wietse In Het Panhuis
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Milena Schönke
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Céline Jouffe
- Institute for Diabetes and Endocrinology (IDE), Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Munich, Germany
| | - Thomas H Devilee
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ricky Siebeler
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Trea C M Streefland
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Hetty C M Sips
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Amanda C M Pronk
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ruben H P Vorderman
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan Bert van Klinken
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands; Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands; Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Nina H Uhlenhaut
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Metabolic Programming, Technical University of Munich School of Life Sciences, Freising, Germany
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
116
|
Jain R, Simcox J. Igniting adipocyte thermogenesis. Cell 2022; 185:941-943. [DOI: 10.1016/j.cell.2022.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 10/18/2022]
|
117
|
Abstract
The role of β-adrenergic receptors (βARs) in adipose tissue to promote lipolysis and the release of fatty acids and nonshivering thermogenesis in brown fat has been studied for so many decades that one would think there is nothing left to discover. With the rediscovery of brown fat in humans and renewed interest in UCP1 and uncoupled mitochondrial respiration, it seems that a review of adipose tissue as an organ, pivotal observations, and the investigators who made them would be instructive to understanding where the field stands now. The discovery of the β3-adrenergic receptor was important for accurately defining the pharmacology of the adipocyte, while the clinical targeting of this receptor for obesity and metabolic disease has had its highs and lows. Many questions still remain about how βARs regulate adipocyte metabolism and the signaling molecules through which they do it.
Collapse
Affiliation(s)
- Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA;
| |
Collapse
|
118
|
Sakers A, De Siqueira MK, Seale P, Villanueva CJ. Adipose-tissue plasticity in health and disease. Cell 2022; 185:419-446. [PMID: 35120662 PMCID: PMC11152570 DOI: 10.1016/j.cell.2021.12.016] [Citation(s) in RCA: 290] [Impact Index Per Article: 145.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022]
Abstract
Adipose tissue, colloquially known as "fat," is an extraordinarily flexible and heterogeneous organ. While historically viewed as a passive site for energy storage, we now appreciate that adipose tissue regulates many aspects of whole-body physiology, including food intake, maintenance of energy levels, insulin sensitivity, body temperature, and immune responses. A crucial property of adipose tissue is its high degree of plasticity. Physiologic stimuli induce dramatic alterations in adipose-tissue metabolism, structure, and phenotype to meet the needs of the organism. Limitations to this plasticity cause diminished or aberrant responses to physiologic cues and drive the progression of cardiometabolic disease along with other pathological consequences of obesity.
Collapse
Affiliation(s)
- Alexander Sakers
- Institute for Diabetes, Obesity & Metabolism, Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Mirian Krystel De Siqueira
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA
| | - Patrick Seale
- Institute for Diabetes, Obesity & Metabolism, Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA.
| | - Claudio J Villanueva
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA.
| |
Collapse
|
119
|
Brownstein AJ, Veliova M, Acin-Perez R, Liesa M, Shirihai OS. ATP-consuming futile cycles as energy dissipating mechanisms to counteract obesity. Rev Endocr Metab Disord 2022; 23:121-131. [PMID: 34741717 PMCID: PMC8873062 DOI: 10.1007/s11154-021-09690-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/24/2021] [Indexed: 12/25/2022]
Abstract
Obesity results from an imbalance in energy homeostasis, whereby excessive energy intake exceeds caloric expenditure. Energy can be dissipated out of an organism by producing heat (thermogenesis), explaining the long-standing interest in exploiting thermogenic processes to counteract obesity. Mitochondrial uncoupling is a process that expends energy by oxidizing nutrients to produce heat, instead of ATP synthesis. Energy can also be dissipated through mechanisms that do not involve mitochondrial uncoupling. Such mechanisms include futile cycles described as metabolic reactions that consume ATP to produce a product from a substrate but then converting the product back into the original substrate, releasing the energy as heat. Energy dissipation driven by cellular ATP demand can be regulated by adjusting the speed and number of futile cycles. Energy consuming futile cycles that are reviewed here are lipolysis/fatty acid re-esterification cycle, creatine/phosphocreatine cycle, and the SERCA-mediated calcium import and export cycle. Their reliance on ATP emphasizes that mitochondrial oxidative function coupled to ATP synthesis, and not just uncoupling, can play a role in thermogenic energy dissipation. Here, we review ATP consuming futile cycles, the evidence for their function in humans, and their potential employment as a strategy to dissipate energy and counteract obesity.
Collapse
Affiliation(s)
- Alexandra J Brownstein
- Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Molecular Cellular Integrative Physiology Interdepartmental Program, University of California, Los Angeles, CA, 90095, USA
| | - Michaela Veliova
- Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Rebeca Acin-Perez
- Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Marc Liesa
- Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Molecular Cellular Integrative Physiology Interdepartmental Program, University of California, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Orian S Shirihai
- Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
- Molecular Cellular Integrative Physiology Interdepartmental Program, University of California, Los Angeles, CA, 90095, USA.
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
120
|
Beulens JWJ, Pinho MGM, Abreu TC, den Braver NR, Lam TM, Huss A, Vlaanderen J, Sonnenschein T, Siddiqui NZ, Yuan Z, Kerckhoffs J, Zhernakova A, Brandao Gois MF, Vermeulen RCH. Environmental risk factors of type 2 diabetes-an exposome approach. Diabetologia 2022; 65:263-274. [PMID: 34792619 DOI: 10.1007/s00125-021-05618-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/07/2021] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes is one of the major chronic diseases accounting for a substantial proportion of disease burden in Western countries. The majority of the burden of type 2 diabetes is attributed to environmental risks and modifiable risk factors such as lifestyle. The environment we live in, and changes to it, can thus contribute substantially to the prevention of type 2 diabetes at a population level. The 'exposome' represents the (measurable) totality of environmental, i.e. nongenetic, drivers of health and disease. The external exposome comprises aspects of the built environment, the social environment, the physico-chemical environment and the lifestyle/food environment. The internal exposome comprises measurements at the epigenetic, transcript, proteome, microbiome or metabolome level to study either the exposures directly, the imprints these exposures leave in the biological system, the potential of the body to combat environmental insults and/or the biology itself. In this review, we describe the evidence for environmental risk factors of type 2 diabetes, focusing on both the general external exposome and imprints of this on the internal exposome. Studies provided established associations of air pollution, residential noise and area-level socioeconomic deprivation with an increased risk of type 2 diabetes, while neighbourhood walkability and green space are consistently associated with a reduced risk of type 2 diabetes. There is little or inconsistent evidence on the contribution of the food environment, other aspects of the social environment and outdoor temperature. These environmental factors are thought to affect type 2 diabetes risk mainly through mechanisms incorporating lifestyle factors such as physical activity or diet, the microbiome, inflammation or chronic stress. To further assess causality of these associations, future studies should focus on investigating the longitudinal effects of our environment (and changes to it) in relation to type 2 diabetes risk and whether these associations are explained by these proposed mechanisms.
Collapse
Affiliation(s)
- Joline W J Beulens
- Department of Epidemiology & Data Science, Amsterdam Public Health, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands.
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Maria G M Pinho
- Department of Epidemiology & Data Science, Amsterdam Public Health, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
| | - Taymara C Abreu
- Department of Epidemiology & Data Science, Amsterdam Public Health, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
| | - Nicole R den Braver
- Department of Epidemiology & Data Science, Amsterdam Public Health, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
| | - Thao M Lam
- Department of Epidemiology & Data Science, Amsterdam Public Health, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
| | - Anke Huss
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Jelle Vlaanderen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Tabea Sonnenschein
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Human Geography and Spatial Planning, Utrecht University, Utrecht, the Netherlands
| | - Noreen Z Siddiqui
- Department of Epidemiology & Data Science, Amsterdam Public Health, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
| | - Zhendong Yuan
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Jules Kerckhoffs
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Milla F Brandao Gois
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Roel C H Vermeulen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
121
|
Xu Z, Chen W, Wang L, You W, Wang Y, Wang Y, Zhao J, Shan T. UCP1 Knockin Induces Lipid Dynamics and Transcriptional Programs in the Skeletal Muscles of Pigs. Front Cell Dev Biol 2022; 9:808095. [PMID: 35096834 PMCID: PMC8790096 DOI: 10.3389/fcell.2021.808095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/07/2021] [Indexed: 02/02/2023] Open
Abstract
Uncoupling protein 1 (UCP1), the hallmark protein responsible for nonshivering thermogenesis in adipose tissue (especially brown adipose tissue) has regained researchers' attention in the context of metabolic disorders following the realization that UCP1 can be activated in adult humans and reconstituted in pigs. Both skeletal muscle and adipose tissue are highly dynamic tissues that interact at the metabolic and hormonal level in response to internal and external stress, and they coordinate in maintaining whole-body metabolic homeostasis. Here, we utilized lipidomics and transcriptomics to identify the altered lipid profiles and regulatory pathways in skeletal muscles from adipocyte-specific UCP1 knock-in (KI) pigs. UCP1 KI changed the contents of glycerophospholipids and acyl carnitines of skeletal muscles. Several metabolic regulatory pathways were more enriched in the UCP1 KI skeletal muscle. Comparison of the transcriptomes of adipose and skeletal muscle suggested that nervous system or chemokine signaling might account for the crosstalk between these two tissues in UCP1 KI pigs. Comparison of the lipid biomarkers from UCP1 KI pigs and other mammals suggested associations between UCP1 KI-induced metabolic alternations and metabolic and muscle dysfunction. Our study reveals the lipid dynamics and transcriptional programs in the skeletal muscle of UCP1 KI pigs and suggests that a network regulates metabolic homeostasis between skeletal muscle and adipose tissue.
Collapse
Affiliation(s)
- Ziye Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Wentao Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Yanfang Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
| | - Jianguo Zhao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
| |
Collapse
|
122
|
Los posibles mecanismos de pardeamiento del tejido adiposo blanco: una diana novedosa para el tratamiento de la obesidad. NUTR HOSP 2022; 39:411-424. [PMID: 35001637 DOI: 10.20960/nh.03852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
The increase of the obesity pandemic worldwide over the last several decades has generated a constant need for the scientific world to develop new possibilities to combat obesity. Since the discovery that brown adipose tissue (BAT) exists in adult humans, and BAT activation contributes to a negative energy balance, much more attention has been focused on the understanding of the molecular switches and their different regulatory mechanisms turning on energy expenditure. Recent insights have revealed that a range of stimuli including cold exposure, physical activity and diet, and critical transcription molecules such as PPARγ, PRDM16, PGC-1α and UCP1, aiming at the induction of BAT activation, could cause the browning of white adipose tissue, thereby dissipating energy and increasing heat production. An increasing number of studies that point to the white adipose tissue (WAT) browning strategies aiming at diet-induced and/or genetically determined obesity have been tested in mouse models as well as in human studies. Findings suggested that browning stimulating drugs have been currently or previously assayed as a therapy against obesity. As PPARα agonists, fibrate drugs effectively reduced plasma triglyceride, increased high-density lipoproteins, and improved glycemic control and heat production in brown adipose tissue, which has been used in the treatment of metabolic disorders. Many kinds of natural products promote white adipose tissue browning, such as alkaloids, flavonoids, terpenoids, and long-chain fatty acids, which can also ameliorate metabolic disorders including obesity, insulin resistance and diabetes. The aim of this review is to summarize the transcriptional regulators as well as the various mediators that have been regarded as potential therapeutic targets in the process of WAT browning.
Collapse
|
123
|
Mills EL, Harmon C, Jedrychowski MP, Xiao H, Gruszczyk AV, Bradshaw GA, Tran N, Garrity R, Laznik-Bogoslavski D, Szpyt J, Prendeville H, Lynch L, Murphy MP, Gygi SP, Spiegelman BM, Chouchani ET. Cysteine 253 of UCP1 regulates energy expenditure and sex-dependent adipose tissue inflammation. Cell Metab 2022; 34:140-157.e8. [PMID: 34861155 PMCID: PMC8732317 DOI: 10.1016/j.cmet.2021.11.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 09/15/2021] [Accepted: 11/08/2021] [Indexed: 01/07/2023]
Abstract
Uncoupling protein 1 (UCP1) is a major regulator of brown and beige adipocyte energy expenditure and metabolic homeostasis. However, the widely employed UCP1 loss-of-function model has recently been shown to have a severe deficiency in the entire electron transport chain of thermogenic fat. As such, the role of UCP1 in metabolic regulation in vivo remains unclear. We recently identified cysteine-253 as a regulatory site on UCP1 that elevates protein activity upon covalent modification. Here, we examine the physiological importance of this site through the generation of a UCP1 cysteine-253-null (UCP1 C253A) mouse, a precise genetic model for selective disruption of UCP1 in vivo. UCP1 C253A mice exhibit significantly compromised thermogenic responses in both males and females but display no measurable effect on fat accumulation in an obesogenic environment. Unexpectedly, we find that a lack of C253 results in adipose tissue redox stress, which drives substantial immune cell infiltration and systemic inflammatory pathology in adipose tissues and liver of male, but not female, mice. Elevation of systemic estrogen reverses this male-specific pathology, providing a basis for protection from inflammation due to loss of UCP1 C253 in females. Together, our results establish the UCP1 C253 activation site as a regulator of acute thermogenesis and sex-dependent tissue inflammation.
Collapse
Affiliation(s)
- Evanna L Mills
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Cathal Harmon
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - Mark P Jedrychowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Haopeng Xiao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Anja V Gruszczyk
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Gary A Bradshaw
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Nhien Tran
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ryan Garrity
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - John Szpyt
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Hannah Prendeville
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Lydia Lynch
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, USA; School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Michael P Murphy
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Edward T Chouchani
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
124
|
Abstract
Brown adipose tissue (BAT) was first identified by Conrad Gessner in 1551, but it was only in 1961 that it was firmly identified as a thermogenic organ. Key developments in the subsequent two decades demonstrated that: (1) BAT is quantitatively important to non-shivering thermogenesis in rodents, (2) uncoupling of oxidative phosphorylation through a mitochondrial proton conductance pathway is the central mechanism by which heat is generated, (3) uncoupling protein-1 is the critical factor regulating proton leakage in BAT mitochondria. Following pivotal studies on cafeteria-fed rats and obese ob/ob mice, BAT was then shown to have a central role in the regulation of energy balance and the etiology of obesity. The application of fluorodeoxyglucose positron emission tomography in the late 2000s confirmed that BAT is present and active in adults, resulting in renewed interest in the tissue in human energetics and obesity. Subsequent studies have demonstrated a broad metabolic role for BAT, the tissue being an important site of glucose disposal and triglyceride clearance, as well as of insulin action. BAT continues to be a potential target for the treatment of obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Paul Trayhurn
- Obesity Biology Unit, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK.
- Clore Laboratory, University of Buckingham, Buckingham, UK.
| |
Collapse
|
125
|
Chen KY, Brychta RJ, Israni NS, Jiang A, Lea HJ, Lentz TN, Pierce AE, Cypess AM. Activating Human Adipose Tissue with the β3-Adrenergic Agonist Mirabegron. Methods Mol Biol 2022; 2448:83-96. [PMID: 35167091 DOI: 10.1007/978-1-0716-2087-8_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
An appealing strategy for treatment of metabolic disease in humans is activation of brown adipose tissue (BAT), a thermogenic organ best visualized through 18F-FDG PET/CT. BAT has been activated to varying degrees by mild cold exposure. However, this approach can cause undesirable stress, and there remains no consensus protocol. Here, we describe standardized methods for both acute and chronic activation of BAT using the orally administered β3-adrenergic receptor (AR) agonist, mirabegron. Acute pharmacological stimulation has enabled quantification of whole-body BAT volume and metabolic activity using PET/CT imaging, and chronic stimulation increases these properties of BAT over time.
Collapse
Affiliation(s)
- Kong Y Chen
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA.
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Robert J Brychta
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nikita S Israni
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alex Jiang
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hannah J Lea
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Taylor N Lentz
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anne E Pierce
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA.
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
126
|
Sorensen J. PET imaging of heart diseases by Acetate. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00209-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
127
|
Jung SM, Le J, Doxsey WG, Haley JA, Park G, Guertin DA, Jang C. Stable Isotope Tracing and Metabolomics to Study In Vivo Brown Adipose Tissue Metabolic Fluxes. Methods Mol Biol 2022; 2448:119-130. [PMID: 35167094 PMCID: PMC9924221 DOI: 10.1007/978-1-0716-2087-8_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Brown adipose tissue (BAT) demonstrates extraordinary metabolic capacity. Previous research using conventional radio tracers reveals that BAT can act as a sink for a diverse menu of nutrients; still, the question of how BAT utilizes these nutrients remains unclear. Recent advances in mass spectrometry (MS) coupled to stable isotope tracing methods have greatly improved our understanding of metabolism in biology. Here, we have developed a BAT-tailored metabolomics and stable isotope tracing protocol using, as an example, the universally labeled 13C-glucose, a key nutrient heavily utilized by BAT. This method enables metabolic roadmaps to be drawn and pathway fluxes to be inferred for each nutrient tracer within BAT and its application could uncover new metabolic pathways not previously appreciated for BAT physiology.
Collapse
Affiliation(s)
- Su Myung Jung
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Biological Sciences, Sungkyunkwan University, Suwon, South Korea
| | - Johnny Le
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Will G Doxsey
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - John A Haley
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Grace Park
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
128
|
Liu X, Zhang Z, Song Y, Xie H, Dong M. An update on brown adipose tissue and obesity intervention: Function, regulation and therapeutic implications. Front Endocrinol (Lausanne) 2022; 13:1065263. [PMID: 36714578 PMCID: PMC9874101 DOI: 10.3389/fendo.2022.1065263] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
Overweight and obesity have become a world-wide problem. However, effective intervention approaches are limited. Brown adipose tissue, which helps maintain body temperature and contributes to thermogenesis, is dependent on uncoupling protein1. Over the last decade, an in-creasing number of studies have found that activating brown adipose tissue and browning of white adipose tissue can protect against obesity and obesity-related metabolic disease. Brown adipose tissue has gradually become an appealing therapeutic target for the prevention and re-versal of obesity. However, some important issues remain unresolved. It is not certain whether increasing brown adipose tissue activity is the cause or effect of body weight loss or what the risks might be for sympathetic nervous system-dependent non-shivering thermogenesis. In this review, we comprehensively summarize approaches to activating brown adipose tissue and/or browning white adipose tissue, such as cold exposure, exercise, and small-molecule treatment. We highlight the functional mechanisms of small-molecule treatment and brown adipose tissue transplantation using batokine, sympathetic nervous system and/or gut microbiome. Finally, we discuss the causality between body weight loss induced by bariatric surgery, exercise, and brown adipose tissue activity.
Collapse
Affiliation(s)
- Xiaomeng Liu
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
- Department of Nutrition and Food Hygiene, College of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhi Zhang
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yajie Song
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Hengchang Xie
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- *Correspondence: Meng Dong, ; Hengchang Xie,
| | - Meng Dong
- Department of Nutrition and Food Hygiene, College of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- *Correspondence: Meng Dong, ; Hengchang Xie,
| |
Collapse
|
129
|
Niclou A, Ocobock C. Weather permitting: Increased seasonal efficiency of nonshivering thermogenesis through brown adipose tissue activation in the winter. Am J Hum Biol 2021; 34:e23716. [PMID: 34942026 DOI: 10.1002/ajhb.23716] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES We investigated seasonal changes in brown adipose tissue (BAT) activation and metabolism in a temperate-climate Albany, NY population. METHODS Data were collected among 58 participants (21 males, 37 females, ages: 18-51) in the summer and 59 participants (23 males, 36 females, ages: 18-63) in the winter in Albany, New York. BAT activity was inferred by comparing metabolic rate, heat dissipation in the supraclavicular area, and respiratory quotient at room temperature and cold exposure. Seasonal variation in BAT was determined by comparing these measurements from summer and winter. RESULTS At mild cold exposure, heat dissipation of the supraclavicular area was significantly greater in the winter compared to summer (p < .001); however, no significant differences were found between seasons in metabolic rate measurements. This suggests BAT activation may be metabolically more efficient in the winter, due to prolonged lower seasonal temperatures relative to summer. Respiratory quotient significantly increased upon mild cold exposure in the winter compared to summer (p < .001). While carbohydrate utilization increased in the winter, fat remained the primary metabolic substrate for BAT activity across both seasons. CONCLUSION The seasonal variations in the effects of nonshivering thermogenesis on metabolic rate and substrate metabolism suggest a buffering of energy expenditure and an increased use of glucose as fuel by BAT as a result of acclimatization to cold in the winter. These findings point towards a potential role of BAT in human whole-body mediated glucose disposal and cold adaptation.
Collapse
Affiliation(s)
- Alexandra Niclou
- Department of Anthropology, University of Notre Dame, Notre Dame, Indiana, USA
| | - Cara Ocobock
- Department of Anthropology, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, Institute for Educational Initiatives, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
130
|
Wade G, McGahee A, Ntambi JM, Simcox J. Lipid Transport in Brown Adipocyte Thermogenesis. Front Physiol 2021; 12:787535. [PMID: 35002769 PMCID: PMC8733649 DOI: 10.3389/fphys.2021.787535] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/02/2021] [Indexed: 12/30/2022] Open
Abstract
Non-shivering thermogenesis is an energy demanding process that primarily occurs in brown and beige adipose tissue. Beyond regulating body temperature, these thermogenic adipocytes regulate systemic glucose and lipid homeostasis. Historically, research on thermogenic adipocytes has focused on glycolytic metabolism due to the discovery of active brown adipose tissue in adult humans through glucose uptake imaging. The importance of lipids in non-shivering thermogenesis has more recently been appreciated. Uptake of circulating lipids into thermogenic adipocytes is necessary for body temperature regulation and whole-body lipid homeostasis. A wide array of circulating lipids contribute to thermogenic potential including free fatty acids, triglycerides, and acylcarnitines. This review will summarize the mechanisms and regulation of lipid uptake into brown adipose tissue including protein-mediated uptake, lipoprotein lipase activity, endocytosis, vesicle packaging, and lipid chaperones. We will also address existing gaps in knowledge for cold induced lipid uptake into thermogenic adipose tissue.
Collapse
Affiliation(s)
| | | | | | - Judith Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
131
|
Thibonnier M, Ghosh S, Blanchard A. Effects of a short-term cold exposure on circulating microRNAs and metabolic parameters in healthy adult subjects. J Cell Mol Med 2021; 26:548-562. [PMID: 34921497 PMCID: PMC8743656 DOI: 10.1111/jcmm.17121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/19/2021] [Accepted: 11/29/2021] [Indexed: 11/28/2022] Open
Abstract
This discovery study investigated in healthy subjects whether a short‐term cold exposure may alter circulating microRNAs and metabolic parameters and if co‐expression networks between these factors could be identified. This open randomized crossover (cold vs no cold exposure) study with blind end‐ point evaluation was conducted at 1 center with 10 healthy adult male volunteers. Wearing a cooling vest perfused at 14°C for 2 h reduced the local skin temperature without triggering shivering, increased norepinephrine and blood pressure while decreasing copeptin, C‐peptide and heart rate. Circulating microRNAs measured before and after wearing the cooling vest twice (4 time points) identified 196 mature microRNAs with excellent reproducibility over 72 h. Significant correlations of microRNA expression with copeptin, norepinephrine and C‐peptide were found. A co‐expression‐based microRNA‐microRNA network, as well as microRNA pairs displaying differential correlation as a function of temperature were also detected. This study demonstrates that circulating miRNAs are differentially expressed and coregulated upon cold exposure in humans, supporting their use as predictive and dynamic biomarkers of cardio‐metabolic disorders.
Collapse
Affiliation(s)
| | - Sujoy Ghosh
- Duke-NUS Medical School, Singapore City, Singapore.,Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Anne Blanchard
- Clinical Investigation Center, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
132
|
Adipose Lipolysis Regulates Cardiac Glucose Uptake and Function in Mice under Cold Stress. Int J Mol Sci 2021; 22:ijms222413361. [PMID: 34948160 PMCID: PMC8703875 DOI: 10.3390/ijms222413361] [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: 10/25/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 02/04/2023] Open
Abstract
The heart primarily uses fatty acids as energy substrates. Adipose lipolysis is a major source of fatty acids, particularly under stress conditions. In this study, we showed that mice with selective inactivation of the lipolytic coactivator comparative gene identification-58 (CGI-58) in adipose tissue (FAT-KO mice), relative to their littermate controls, had lower circulating FA levels in the fed and fasted states due to impaired adipose lipolysis. They preferentially utilized carbohydrates as energy fuels and were more insulin sensitive and glucose tolerant. Under cold stress, FAT-KO versus control mice had >10-fold increases in glucose uptake in the hearts but no increases in other tissues examined. Plasma concentrations of atrial natriuretic peptide and cardiac mRNAs for atrial and brain-type natriuretic peptides, two sensitive markers of cardiac remodeling, were also elevated. After one week of cold exposure, FAT-KO mice showed reduced cardiac expression of several mitochondrial oxidative phosphorylation proteins. After one month of cold exposure, hearts of these animals showed depressed functions, reduced SERCA2 protein, and increased proteins for MHC-β, collagen I proteins, Glut1, Glut4 and phospho-AMPK. Thus, CGI-58-dependent adipose lipolysis critically regulates cardiac metabolism and function, especially during cold adaptation. The adipose-heart axis may be targeted for the management of cardiac dysfunction.
Collapse
|
133
|
Nishi M, Ogata T, Kobayakawa K, Kobayakawa R, Matsuo T, Cannistraci CV, Tomita S, Taminishi S, Suga T, Kitani T, Higuchi Y, Sakamoto A, Tsuji Y, Soga T, Matoba S. Energy-sparing by 2-methyl-2-thiazoline protects heart from ischaemia/reperfusion injury. ESC Heart Fail 2021; 9:428-441. [PMID: 34854235 PMCID: PMC8787978 DOI: 10.1002/ehf2.13732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 10/13/2021] [Accepted: 11/11/2021] [Indexed: 11/06/2022] Open
Abstract
AIMS Cardiac ischaemia/reperfusion (I/R) injury remains a critical issue in the therapeutic management of ischaemic heart failure. Although mild hypothermia has a protective effect on cardiac I/R injury, more rapid and safe methods that can obtain similar results to hypothermia therapy are required. 2-Methyl-2-thiazoline (2MT), an innate fear inducer, causes mild hypothermia resulting in resistance to critical hypoxia in cutaneous or cerebral I/R injury. The aim of this study is to demonstrate the protective effect of systemically administered 2MT on cardiac I/R injury and to elucidate the mechanism underlying this effect. METHODS AND RESULTS A single subcutaneous injection of 2MT (50 mg/kg) was given prior to reperfusion of the I/R injured 10 week-old male mouse heart and its efficacy was evaluated 24 h after the ligation of the left anterior descending coronary artery. 2MT preserved left ventricular systolic function following I/R injury (ejection fraction, %: control 37.9 ± 6.7, 2MT 54.1 ± 6.4, P < 0.01). 2MT also decreased infarct size (infarct size/ischaemic area at risk, %: control 48.3 ± 12.1, 2MT 25.6 ± 4.2, P < 0.05) and serum cardiac troponin levels (ng/mL: control 8.9 ± 1.1, 2MT 1.9 ± 0.1, P < 0.01) after I/R. Moreover, 2MT reduced the oxidative stress-exposed area within the heart (%: control 25.3 ± 4.7, 2MT 10.8 ± 1.4, P < 0.01). These results were supported by microarray analysis of the mouse hearts. 2MT induced a transient, mild decrease in core body temperature (°C: -2.4 ± 1.4), which gradually recovered over several hours. Metabolome analysis of the mouse hearts suggested that 2MT minimized energy metabolism towards suppressing oxidative stress. Furthermore, 18F-fluorodeoxyglucose-positron emission tomography/computed tomography imaging revealed that 2MT reduced the activity of brown adipose tissue (standardized uptake value: control 24.3 ± 6.4, 2MT 18.4 ± 5.8, P < 0.05). 2MT also inhibited mitochondrial respiration and glycolysis in rat cardiomyoblasts. CONCLUSIONS We identified the cardioprotective effect of systemically administered 2MT on cardiac I/R injury by sparing energy metabolism with reversible hypothermia. Our results highlight the potential of drug-induced hypothermia therapy as an adjunct to coronary intervention in severe ischaemic heart disease.
Collapse
Affiliation(s)
- Masahiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Ko Kobayakawa
- Functional Neuroscience Lab, Kansai Medical University, Hirakata, Japan
| | - Reiko Kobayakawa
- Functional Neuroscience Lab, Kansai Medical University, Hirakata, Japan
| | - Tomohiko Matsuo
- Functional Neuroscience Lab, Kansai Medical University, Hirakata, Japan
| | - Carlo Vittorio Cannistraci
- Center for Complex Network Intelligence (CCNI), Tsinghua Laboratory of Brain and Intelligence (THBI), Department of Computer Science, Department of Biomedical Engineering, Tsinghua University, China.,Center for Systems Biology Dresden (CSBD), Dresden, Germany
| | - Shinya Tomita
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shunta Taminishi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takaomi Suga
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoya Kitani
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yusuke Higuchi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akira Sakamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yumika Tsuji
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
134
|
Tournissac M, Leclerc M, Valentin-Escalera J, Vandal M, Bosoi CR, Planel E, Calon F. Metabolic determinants of Alzheimer's disease: A focus on thermoregulation. Ageing Res Rev 2021; 72:101462. [PMID: 34534683 DOI: 10.1016/j.arr.2021.101462] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/09/2021] [Accepted: 09/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a complex age-related neurodegenerative disease, associated with central and peripheral metabolic anomalies, such as impaired glucose utilization and insulin resistance. These observations led to a considerable interest not only in lifestyle-related interventions, but also in repurposing insulin and other anti-diabetic drugs to prevent or treat dementia. Body temperature is the oldest known metabolic readout and mechanisms underlying its maintenance fail in the elderly, when the incidence of AD rises. This raises the possibility that an age-associated thermoregulatory deficit contributes to energy failure underlying AD pathogenesis. Brown adipose tissue (BAT) plays a central role in thermogenesis and maintenance of body temperature. In recent years, the modulation of BAT activity has been increasingly demonstrated to regulate energy expenditure, insulin sensitivity and glucose utilization, which could also provide benefits for AD. Here, we review the evidence linking thermoregulation, BAT and insulin-related metabolic defects with AD, and we propose mechanisms through which correcting thermoregulatory impairments could slow the progression and delay the onset of AD.
Collapse
|
135
|
Li F, Jing J, Movahed M, Cui X, Cao Q, Wu R, Chen Z, Yu L, Pan Y, Shi H, Shi H, Xue B. Epigenetic interaction between UTX and DNMT1 regulates diet-induced myogenic remodeling in brown fat. Nat Commun 2021; 12:6838. [PMID: 34824202 PMCID: PMC8617140 DOI: 10.1038/s41467-021-27141-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/05/2021] [Indexed: 02/04/2023] Open
Abstract
Brown adipocytes share the same developmental origin with skeletal muscle. Here we find that a brown adipocyte-to-myocyte remodeling also exists in mature brown adipocytes, and is induced by prolonged high fat diet (HFD) feeding, leading to brown fat dysfunction. This process is regulated by the interaction of epigenetic pathways involving histone and DNA methylation. In mature brown adipocytes, the histone demethylase UTX maintains persistent demethylation of the repressive mark H3K27me3 at Prdm16 promoter, leading to high Prdm16 expression. PRDM16 then recruits DNA methyltransferase DNMT1 to Myod1 promoter, causing Myod1 promoter hypermethylation and suppressing its expression. The interaction between PRDM16 and DNMT1 coordinately serves to maintain brown adipocyte identity while repressing myogenic remodeling in mature brown adipocytes, thus promoting their active brown adipocyte thermogenic function. Suppressing this interaction by HFD feeding induces brown adipocyte-to-myocyte remodeling, which limits brown adipocyte thermogenic capacity and compromises diet-induced thermogenesis, leading to the development of obesity.
Collapse
Affiliation(s)
- Fenfen Li
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Jia Jing
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Miranda Movahed
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Xin Cui
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Qiang Cao
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Rui Wu
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Ziyue Chen
- grid.256304.60000 0004 1936 7400Department of Computer Science, Georgia State University, Atlanta, GA 30303 USA
| | - Liqing Yu
- grid.411024.20000 0001 2175 4264Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Yi Pan
- grid.256304.60000 0004 1936 7400Department of Computer Science, Georgia State University, Atlanta, GA 30303 USA ,grid.458489.c0000 0001 0483 7922Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 P.R. China
| | - Huidong Shi
- grid.410427.40000 0001 2284 9329Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912 USA ,grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912 USA
| | - Hang Shi
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Bingzhong Xue
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| |
Collapse
|
136
|
Augmented CCL5/CCR5 signaling in brown adipose tissue inhibits adaptive thermogenesis and worsens insulin resistance in obesity. Clin Sci (Lond) 2021; 136:121-137. [PMID: 34821367 DOI: 10.1042/cs20210959] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022]
Abstract
Chemokine (C-C motif) ligand 5 (CCL5) and CCR5, one of its receptors have been reported to be highly expressed in white adipose tissue (WAT) and are associated with the progression of inflammation and the development of insulin resistance in obese humans and mice. However, the role of CCL5/CCR5 signaling in obesity-associated dysregulation of energy metabolism remains unclear. Here, we demonstrate that global CCL5/CCR5 double knockout (DKO) mice have higher cold stress-induced energy expenditure and thermogenic function in BAT than wild-type (WT) mice. DKO mice have higher cold stress-induced energy expenditure and thermogenic function in BAT than wild-type mice. KEGG pathway analysis indicated that deletion of CCL5/CCR5 further facilitated the cold-induced expression of genes related to oxidative phosphorylation and lipid metabolic pathways. In primary brown adipocytes of DKO mice, the augmentation of CL-316243-stimulated thermogenic and lipolysis responses was reversed by co-treatment with AMPKα1 and α2 siRNA. Overexpression of BAT CCL5/CCR5 genes by local lentivirus injection in WT mice suppressed cold stress-induced lipolytic processes and thermogenic activities. In contrast, knockdown of BAT CCL5/CCR5 signaling further upregulated AMPK phosphorylation as well as thermogenic and lipolysis responses to chronic adrenergic stimuli and subsequently decreased level of body weight gain. Chronic knockdown of BAT CCL5/CCR5 signaling improved HFD-induced insulin resistance in WT mice. It is suggested that obesity-induced augmentation of AT CCL5/CCR5 signaling could, at least in part, suppress energy expenditure and adaptive thermogenesis by inhibiting AMPK-mediated lipolysis and oxidative metabolism in thermogenic AT to exacerbate the development of obesity and insulin resistance.
Collapse
|
137
|
Law JM, Morris DE, Robinson LJ, Symonds ME, Budge H. Semi-automated analysis of supraclavicular thermal images increases speed of brown adipose tissue analysis without increasing variation in results. Curr Res Physiol 2021; 4:177-182. [PMID: 34746836 PMCID: PMC8562194 DOI: 10.1016/j.crphys.2021.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/22/2021] [Accepted: 07/07/2021] [Indexed: 12/03/2022] Open
Abstract
Interest in brown adipose tissue remains high a decade after it was determined to be present outside of the neonatal period. In vivo imaging, however, has remained a challenge due to the lack of a imaging modality suitable for large healthy-volunteer studies, post-prandial investigations and vulnerable groups, such as children. Infrared thermography is increasingly accepted as a valid, non-invasive and flexible alternative but there is a wide approach to analysis between different groups. Defining the region of interest with anatomical borders rather than using a simple polygon may have advantages in terms of consistency but makes image analysis slower, limiting some applications. Our novel semi-automated method, using a custom-built graphical user interface, allows an 86% improvement in speed of image analysis (54.9 (38.3–71.4) seconds/image) without increases in variation between analysers or with repeated analysis. The improved efficiency demonstrated makes feasible larger studies, longer imaging periods or increased image acquisition frequency, providing an opportunity to study novel features of brown adipose tissue function. Brown adipose tissue is a key heat-generating tissue but is difficult to measure. Thermal imaging can measure brown adipose tissue response without radiation. A semi-automated approach increases image analysis efficiency. Thermal video analysis and imaging over longer periods is now feasible.
Collapse
Affiliation(s)
- James M Law
- Early Life Research Unit, Division of Child Health, Obstetrics & Gynaecology, University of Nottingham, United Kingdom
| | - David E Morris
- Bioengineering Research Group, Faculty of Engineering, University of Nottingham, United Kingdom
| | - Lindsay J Robinson
- Early Life Research Unit, Division of Child Health, Obstetrics & Gynaecology, University of Nottingham, United Kingdom
| | - Michael E Symonds
- Early Life Research Unit, Division of Child Health, Obstetrics & Gynaecology, University of Nottingham, United Kingdom.,Nottingham Digestive Disease Centre and Biomedical Research Centre, School of Medicine, University of Nottingham, NG7 2UH, United Kingdom
| | - Helen Budge
- Early Life Research Unit, Division of Child Health, Obstetrics & Gynaecology, University of Nottingham, United Kingdom
| |
Collapse
|
138
|
Human Brown Adipose Tissue and Metabolic Health: Potential for Therapeutic Avenues. Cells 2021; 10:cells10113030. [PMID: 34831253 PMCID: PMC8616549 DOI: 10.3390/cells10113030] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/31/2022] Open
Abstract
Obesity-associated metabolic abnormalities comprise a cluster of conditions including dyslipidemia, insulin resistance, diabetes and cardiovascular diseases that has affected more than 650 million people all over the globe. Obesity results from the accumulation of white adipose tissues mainly due to the chronic imbalance of energy intake and energy expenditure. A variety of approaches to treat or prevent obesity, including lifestyle interventions, surgical weight loss procedures and pharmacological approaches to reduce energy intake and increase energy expenditure have failed to substantially decrease the prevalence of obesity. Brown adipose tissue (BAT), the primary source of thermogenesis in infants and small mammals may represent a promising therapeutic target to treat obesity by promoting energy expenditure through non-shivering thermogenesis mediated by mitochondrial uncoupling protein 1 (UCP1). Since the confirmation of functional BAT in adult humans by several groups, approximately a decade ago, and its association with a favorable metabolic phenotype, intense interest on the significance of BAT in adult human physiology and metabolic health has emerged within the scientific community to explore its therapeutic potential for the treatment of obesity and metabolic diseases. A substantially decreased BAT activity in individuals with obesity indicates a role for BAT in the setting of human obesity. On the other hand, BAT mass and its prevalence correlate with lower body mass index (BMI), decreased age and lower glucose levels, leading to a lower incidence of cardio-metabolic diseases. The increased cold exposure in adult humans with undetectable BAT was associated with decreased body fat mass and increased insulin sensitivity. A deeper understanding of the role of BAT in human metabolic health and its interrelationship with body fat distribution and deciphering proper strategies to increase energy expenditure, by either increasing functional BAT mass or inducing white adipose browning, holds the promise for possible therapeutic avenues for the treatment of obesity and associated metabolic disorders.
Collapse
|
139
|
Yuko OO, Saito M. Brown Fat as a Regulator of Systemic Metabolism beyond Thermogenesis. Diabetes Metab J 2021; 45:840-852. [PMID: 34176254 PMCID: PMC8640153 DOI: 10.4093/dmj.2020.0291] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/26/2021] [Indexed: 12/01/2022] Open
Abstract
Brown adipose tissue (BAT) is a specialized tissue for nonshivering thermogenesis to dissipate energy as heat. Although BAT research has long been limited mostly in small rodents, the rediscovery of metabolically active BAT in adult humans has dramatically promoted the translational studies on BAT in health and diseases. Moreover, several remarkable advancements have been made in brown fat biology over the past decade: The molecular and functional analyses of inducible thermogenic adipocytes (socalled beige adipocytes) arising from a developmentally different lineage from classical brown adipocytes have been accelerated. In addition to a well-established thermogenic activity of uncoupling protein 1 (UCP1), several alternative thermogenic mechanisms have been discovered, particularly in beige adipocytes. It has become clear that BAT influences other peripheral tissues and controls their functions and systemic homeostasis of energy and metabolic substrates, suggesting BAT as a metabolic regulator, other than for thermogenesis. This notion is supported by discovering that various paracrine and endocrine factors are secreted from BAT. We review the current understanding of BAT pathophysiology, particularly focusing on its role as a metabolic regulator in small rodents and also in humans.
Collapse
Affiliation(s)
| | - Masayuki Saito
- Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Nutrition, Tenshi College, Sapporo, Japan
- Corresponding author: Masayuki Saito https://orcid.org/0000-0002-3058-3003 Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan E-mail:
| |
Collapse
|
140
|
Role of RAGE in obesity-induced adipose tissue inflammation and insulin resistance. Cell Death Discov 2021; 7:305. [PMID: 34686659 PMCID: PMC8536716 DOI: 10.1038/s41420-021-00711-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/22/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
Obesity is known to be associated with adipose tissue inflammation and insulin resistance. Importantly, in obesity, the accumulation of proinflammatory macrophages in adipose tissue correlates with insulin resistance. We hypothesized that the receptor for advanced glycation end products (RAGE) and associated ligands are involved in adipose tissue insulin resistance, and that the activation of the AGE–RAGE axis plays an important role in obesity-associated inflammation. C57BL/6J mice (WT) and RAGE deficient (RAGE−/−) mice were fed a high fat diet (HFD) and subjected to glucose and insulin tolerance tests. Epdidymal adipose tissue (eAT) was collected and adipose stromal vascular cells isolated using flow cytometry. Visceral adipose tissue macrophage polarization was assessed by quantitative real time PCR. Immunoblotting was performed to evaluate the insulin signaling in adipose tissues. In additional studies, cell trafficking was assessed by injecting labeled blood monocytes into recipient mice. RAGE−/− mice displayed improved insulin sensitivity and glucose tolerance, accompanied by decreased body weight and eAT mass. Exogenous methylglyoxal (MGO) impaired insulin-stimulated AKT signaling in adipose tissues from WT mice fed a normal chow diet, but not in RAGE−/− mice. In contrast, in obese mice, treatment with MGO did not reduce insulin-induced phosphorylation of AKT in WT-HFD mice. Moreover, insulin-induced AKT phosphorylation was found to be impaired in adipose tissue from RAGE−/−-HFD mice. RAGE−/− mice displayed improved inflammatory profiles and evidence for increased adipose tissue browning. This observation is consistent with the finding of reduced plasma levels of FFA, glycerol, IL-6, and leptin in RAGE−/− mice compared to WT mice. Collectively the data demonstrate that RAGE-mediated adipose tissue inflammation and insulin-signaling are potentially important mechanisms that contribute to the development of obesity-associated insulin resistance.
Collapse
|
141
|
Acín-Perez R, Petcherski A, Veliova M, Benador IY, Assali EA, Colleluori G, Cinti S, Brownstein AJ, Baghdasarian S, Livhits MJ, Yeh MW, Krishnan KC, Vergnes L, Winn NC, Padilla J, Liesa M, Sacks HS, Shirihai OS. Recruitment and remodeling of peridroplet mitochondria in human adipose tissue. Redox Biol 2021; 46:102087. [PMID: 34411987 PMCID: PMC8377484 DOI: 10.1016/j.redox.2021.102087] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 01/31/2023] Open
Abstract
Beige adipocyte mitochondria contribute to thermogenesis by uncoupling and by ATP-consuming futile cycles. Since uncoupling may inhibit ATP synthesis, it is expected that expenditure through ATP synthesis is segregated to a disparate population of mitochondria. Recent studies in mouse brown adipocytes identified peridroplet mitochondria (PDM) as having greater ATP synthesis and pyruvate oxidation capacities, while cytoplasmic mitochondria have increased fatty acid oxidation and uncoupling capacities. However, the occurrence of PDM in humans and the processes that result in their expansion have not been elucidated. Here, we describe a novel high-throughput assay to quantify PDM that is successfully applied to white adipose tissue from mice and humans. Using this approach, we found that PDM content varies between white and brown fat in both species. We used adipose tissue from pheochromocytoma (Pheo) patients as a model of white adipose tissue browning, which is characterized by an increase in the capacity for energy expenditure. In contrast with control subjects, PDM content was robustly increased in the periadrenal fat of Pheo patients. Remarkably, bioenergetic changes associated with browning were primarily localized to PDM compared to cytoplasmic mitochondria (CM). PDM isolated from periadrenal fat of Pheo patients had increased ATP-linked respiration, Complex IV content and activity, and maximal respiratory capacity. We found similar changes in a mouse model of re-browning where PDM content in whitened brown adipose tissue was increased upon re-browning induced by decreased housing temperature. Taken together, this study demonstrates the existence of PDM as a separate functional entity in humans and that browning in both mice and humans is associated with a robust expansion of peri-droplet mitochondria characterized by increased ATP synthesis linked respiration.
Collapse
Affiliation(s)
- Rebeca Acín-Perez
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Anton Petcherski
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Michaela Veliova
- Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Ilan Y Benador
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Nutrition and Metabolism, Graduate Medical Sciences, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Essam A Assali
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Department of Clinical Biochemistry, School of Medicine, Ben Gurion University of The Negev, Beer-Sheva, Israel
| | - Georgia Colleluori
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Ancona, 60020, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Ancona, 60020, Italy
| | - Alexandra J Brownstein
- Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Molecular Cellular Integrative Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Siyouneh Baghdasarian
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Masha J Livhits
- Section of Endocrine Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Michael W Yeh
- Section of Endocrine Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Karthickeyan Chella Krishnan
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, Los Angeles, CA, USA; Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, OH, USA
| | - Laurent Vergnes
- Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Nathan C Winn
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA; Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jaume Padilla
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Marc Liesa
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA; Molecular Cellular Integrative Physiology, University of California, Los Angeles, CA, 90095, USA.
| | - Harold S Sacks
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Endocrine and Diabetes Division, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA.
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA; Nutrition and Metabolism, Graduate Medical Sciences, Boston University School of Medicine, Boston, MA, 02118, USA; Molecular Cellular Integrative Physiology, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
142
|
Flanagan EW, Altazan AD, Carmichael OT, Hu HH, Redman LM. Practical application of in vivo MRI-based brown adipose tissue measurements in infants. Obesity (Silver Spring) 2021; 29:1676-1683. [PMID: 34553508 PMCID: PMC9115839 DOI: 10.1002/oby.23237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The role of brown adipose tissue (BAT) in infant metabolism remains poorly understood, primarily because of the inherent limitation of positron emission tomography/computed tomography imaging to measure BAT, which is not suitable for infants. The aims of this method development study were to assess the feasibility, intra-rater reliability, interscan repeatability, and physiological relevance of measuring BAT in infants using magnetic resonance imaging (MRI). METHODS A total of 10 nonsedated infants (mean age, 22.6 [1.3] days old) completed two 3-T MRI exams using chemical-shift-encoded water-fat scans 6.2 (2.8) days apart. Candidate BAT voxels in the supraclavicular region were identified based on fat signal fraction (FSF). The volumes of BAT depots were manually traced, and FSF was calculated. Whole-body fat mass was determined using dual-energy x-ray absorptiometry. RESULTS Images were successfully obtained from 19 of 20 (95%) attempted scans. The mean BAT volume was 5.41 (SD 1.1) cm3 , and the mean FSF was 16.41% (SD 3.3%). Intra-rater analysis showed good reliability with no systemic bias (proportional bias for volume: p = 0.19; FSF: p = 0.30). Test-retest for interscan repeatability was good (intraclass correlation coefficients for volume: 0.92, p = 0.001 and intraclass correlation coefficients for FSF: 0.93, p < 0.001). FSF was inversely related to fat-free mass (r = -0.69, p = 0.03). CONCLUSIONS This method development study supports the use of MRI to obtain reliable and quantitative measurements of BAT volume in infants.
Collapse
Affiliation(s)
- Emily W Flanagan
- Pennington Biomedical Research Center, Baton Rouge, Louisana, USA
| | - Abby D Altazan
- Pennington Biomedical Research Center, Baton Rouge, Louisana, USA
| | | | - Houchun H Hu
- Hyperfine Research, Inc., Guilford, Connecticut, USA
| | - Leanne M Redman
- Pennington Biomedical Research Center, Baton Rouge, Louisana, USA
| |
Collapse
|
143
|
Yan S, Kumari M, Xiao H, Jacobs C, Kochumon S, Jedrychowski M, Chouchani E, Ahmad R, Rosen ED. IRF3 reduces adipose thermogenesis via ISG15-mediated reprogramming of glycolysis. J Clin Invest 2021; 131:144888. [PMID: 33571167 DOI: 10.1172/jci144888] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/10/2021] [Indexed: 12/21/2022] Open
Abstract
Adipose thermogenesis is repressed in obesity, reducing the homeostatic capacity to compensate for chronic overnutrition. Inflammation inhibits adipose thermogenesis, but little is known about how this occurs. Here we showed that the innate immune transcription factor IRF3 is a strong repressor of thermogenic gene expression and oxygen consumption in adipocytes. IRF3 achieved this by driving expression of the ubiquitin-like modifier ISG15, which became covalently attached to glycolytic enzymes, thus reducing their function and decreasing lactate production. Lactate repletion was able to restore thermogenic gene expression, even when the IRF3/ISG15 axis was activated. Mice lacking ISG15 phenocopied mice lacking IRF3 in adipocytes, as both had elevated energy expenditure and were resistant to diet-induced obesity. These studies provide a deep mechanistic understanding of how the chronic inflammatory milieu of adipose tissue in obesity prevents thermogenic compensation for overnutrition.
Collapse
Affiliation(s)
- Shuai Yan
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Manju Kumari
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.,Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Haopeng Xiao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher Jacobs
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Shihab Kochumon
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Mark Jedrychowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Edward Chouchani
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Evan D Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
144
|
Ahmed BA, Ong FJ, Barra NG, Blondin DP, Gunn E, Oreskovich SM, Szamosi JC, Syed SA, Hutchings EK, Konyer NB, Singh NP, Yabut JM, Desjardins EM, Anhê FF, Foley KP, Holloway AC, Noseworthy MD, Haman F, Carpentier AC, Surette MG, Schertzer JD, Punthakee Z, Steinberg GR, Morrison KM. Lower brown adipose tissue activity is associated with non-alcoholic fatty liver disease but not changes in the gut microbiota. Cell Rep Med 2021; 2:100397. [PMID: 34622234 PMCID: PMC8484690 DOI: 10.1016/j.xcrm.2021.100397] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/25/2021] [Accepted: 08/18/2021] [Indexed: 12/18/2022]
Abstract
In rodents, lower brown adipose tissue (BAT) activity is associated with greater liver steatosis and changes in the gut microbiome. However, little is known about these relationships in humans. In adults (n = 60), we assessed hepatic fat and cold-stimulated BAT activity using magnetic resonance imaging and the gut microbiota with 16S sequencing. We transplanted gnotobiotic mice with feces from humans to assess the transferability of BAT activity through the microbiota. Individuals with NAFLD (n = 29) have lower BAT activity than those without, and BAT activity is inversely related to hepatic fat content. BAT activity is not related to the characteristics of the fecal microbiota and is not transmissible through fecal transplantation to mice. Thus, low BAT activity is associated with higher hepatic fat accumulation in human adults, but this does not appear to have been mediated through the gut microbiota.
Collapse
Affiliation(s)
- Basma A. Ahmed
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Frank J. Ong
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Nicole G. Barra
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Denis P. Blondin
- Faculty of Medicine and Health Sciences, Department of Medicine, Division of Neurology, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Elizabeth Gunn
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Stephan M. Oreskovich
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jake C. Szamosi
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
- Farncombe Metagenomics Facility, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Saad A. Syed
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Emily K. Hutchings
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Norman B. Konyer
- Imaging Research Centre, St. Joseph’s Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Nina P. Singh
- Department of Radiology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Julian M. Yabut
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Eric M. Desjardins
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Fernando F. Anhê
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Kevin P. Foley
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Alison C. Holloway
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Michael D. Noseworthy
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Imaging Research Centre, St. Joseph’s Healthcare, Hamilton, ON L8N 4A6, Canada
- Department of Radiology, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Electrical and Computer Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
- School of Biomedical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Francois Haman
- Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Andre C. Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Michael G. Surette
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jonathan D. Schertzer
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Zubin Punthakee
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Gregory R. Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Katherine M. Morrison
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
145
|
Greenfield AM, Charkoudian N, Alba BK. Influences of ovarian hormones on physiological responses to cold in women. Temperature (Austin) 2021; 9:23-45. [DOI: 10.1080/23328940.2021.1953688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Andrew Martin Greenfield
- Thermal & Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
- Oak Ridge Institute of Science and Education, Belcamp, MD, USA
| | - Nisha Charkoudian
- Thermal & Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Billie Katherine Alba
- Thermal & Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| |
Collapse
|
146
|
Ocobock C, Soppela P, Turunen M, Stenbäck V, Herzig KH, Rimbach R, Pontzer H. Reindeer herders from subarctic Finland exhibit high total energy expenditure and low energy intake during the autumn herd roundup. Am J Hum Biol 2021; 34:e23676. [PMID: 34520587 DOI: 10.1002/ajhb.23676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE High levels of total energy expenditure (TEE, kcal/day) have been documented among numerous human populations such as tropical climate horticulturalists and high-altitude agriculturalists. However, less work has been conducted among highly physically active cold climate populations. METHODS In October 2018, TEE was measured using the doubly labeled water (TEEDLW , N = 10) and flex-heart rate methods (TEEHR , N = 24) for 6-14 days among reindeer herders (20-62 years) in northern Finland during an especially physically demanding, but not seasonally representative, period of the year for herders-the annual reindeer herd roundup. Self-reported dietary intake was also collected during TEE measurement periods. TEE was then compared to that of hunter gatherer, farming, and market economies. RESULTS During the herd roundup, herders expended a mean of 4183 ± 949 kcal/day as measured by the DLW method, which was not significantly different from TEEHR . Mean caloric intake was 1718 ± 709 kcal/day, and was significantly lower than TEEDLW and TEEHR (p < .001). Herder TEEDLW was significantly higher than that of hunter gatherer (p = .0014) and market (p < .0014) economy populations; however, herder TEEDLW was not different from that of farming populations (p = .91). CONCLUSION High TEE and low caloric intake among herders reflect the extreme demands placed on herders during the annual herd round up. Although TEEDLW was similar between cold climate herders and hot climate farming populations, there are likely differences in how that TEE is comprised, reflecting the local ecologies of these populations.
Collapse
Affiliation(s)
- Cara Ocobock
- Department of Anthropology, University of Notre Dame, Notre Dame, Indiana, USA.,Eck Institute for Global Health, Institute for Educational Initiatives, University of Notre Dame, Notre Dame, Indiana, USA
| | - Päivi Soppela
- University of Lapland, Arctic Centre, Rovaniemi, Finland
| | - Minna Turunen
- University of Lapland, Arctic Centre, Rovaniemi, Finland
| | - Ville Stenbäck
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu and Oulu University Hospital, Oulu, Finland.,Biocenter Oulu, Oulu, Finland
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu and Oulu University Hospital, Oulu, Finland.,Institute of Pediatrics, Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, Poznan, Poland
| | - Rebecca Rimbach
- Department of Evolutionary Anthropology, Duke University, Durham, North Carolina, USA.,School of Animal, Plant & Environmental Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Herman Pontzer
- Department of Evolutionary Anthropology, Duke University, Durham, North Carolina, USA.,Duke Global Health Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
147
|
Metabolomic Analysis Reveals Changes in Plasma Metabolites in Response to Acute Cold Stress and Their Relationships to Metabolic Health in Cold-Acclimatized Humans. Metabolites 2021; 11:metabo11090619. [PMID: 34564435 PMCID: PMC8468536 DOI: 10.3390/metabo11090619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/08/2021] [Indexed: 11/29/2022] Open
Abstract
Cold exposure results in activation of metabolic processes required for fueling thermogenesis, potentially promoting improved metabolic health. However, the metabolic complexity underlying this process is not completely understood. We aimed to analyze changes in plasma metabolites related to acute cold exposure and their relationship to cold-acclimatization level and metabolic health in cold-acclimatized humans. Blood samples were obtained before and acutely after 10–15 min of ice-water swimming (<5 °C) from 14 ice-water swimmers. Using mass spectrometry, 973 plasma metabolites were measured. Ice-water swimming induced acute changes in 70 metabolites. Pathways related to amino acid metabolism were the most cold-affected and cold-induced changes in several amino acids correlated with cold-acclimatization level and/or metabolic health markers, including atherogenic lipid profile or insulin resistance. Metabolites correlating with cold-acclimatization level were enriched in the linoleic/α-linolenic acid metabolic pathway. N-lactoyl-tryptophan correlated with both cold-acclimatization level and cold-induced changes in thyroid and parathyroid hormones. Acute cold stress in cold-acclimatized humans induces changes in plasma metabolome that involve amino acids metabolism, while the linoleic and α-linolenic acid metabolism pathway seems to be affected by regular cold exposure. Metabolites related to metabolic health, thermogenic hormonal regulators and acclimatization level might represent prospective molecular factors important in metabolic adaptations to regular cold exposure.
Collapse
|
148
|
Role of Brown and Beige Adipose Tissues in Seasonal Adaptation in the Raccoon Dog ( Nyctereutes procyonoides). Int J Mol Sci 2021; 22:ijms22179623. [PMID: 34502532 PMCID: PMC8431801 DOI: 10.3390/ijms22179623] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/05/2021] [Accepted: 08/31/2021] [Indexed: 01/18/2023] Open
Abstract
Brown adipose tissue (BAT) expresses uncoupling protein-1 (UCP1), which enables energy to be exerted towards needed thermogenesis. Beige adipocytes are precursor cells interspersed among white adipose tissue (WAT) that possess similar UCP1 activity and capacity for thermogenesis. The raccoon dog (Nyctereutes procyonoides) is a canid species that utilizes seasonal obesity to survive periods of food shortage in climate zones with cold winters. The potential to recruit a part of the abundant WAT storages as beige adipocytes for UCP1-dependent thermogenesis was investigated in vitro by treating raccoon dog adipocytes with different browning inducing factors. In vivo positron emission tomography/computed tomography (PET/CT) imaging with the glucose analog 18F-FDG showed that BAT was not detected in the adult raccoon dog during the winter season. In addition, UCP1 expression was not changed in response to chronic treatments with browning inducing factors in adipocyte cultures. Our results demonstrated that most likely the raccoon dog endures cold weather without the induction of BAT or recruitment of beige adipocytes for heat production. Its thick fur coat, insulating fat, and muscle shivering seem to provide the adequate heat needed for surviving the winter.
Collapse
|
149
|
Halbgebauer D, Roos J, Funcke JB, Neubauer H, Hamilton BS, Simon E, Amri EZ, Debatin KM, Wabitsch M, Fischer-Posovszky P, Tews D. Latent TGFβ-binding proteins regulate UCP1 expression and function via TGFβ2. Mol Metab 2021; 53:101336. [PMID: 34481123 PMCID: PMC8456047 DOI: 10.1016/j.molmet.2021.101336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 12/26/2022] Open
Abstract
Objective Activation of brown adipose tissue (BAT) in humans has been proposed as a new treatment approach for combating obesity and its associated diseases, as BAT participates in the regulation of energy homeostasis as well as glucose and lipid metabolism. Genetic contributors driving brown adipogenesis in humans have not been fully understood. Methods Profiling the gene expression of progenitor cells from subcutaneous and deep neck adipose tissue, we discovered new secreted factors with potential regulatory roles in white and brown adipogenesis. Among these, members of the latent transforming growth factor beta-binding protein (LTBP) family were highly expressed in brown compared to white adipocyte progenitor cells, suggesting that these proteins are capable of promoting brown adipogenesis. To investigate this potential, we used CRISPR/Cas9 to generate LTBP-deficient human preadipocytes. Results We demonstrate that LTBP2 and LTBP3 deficiency does not affect adipogenic differentiation, but diminishes UCP1 expression and function in the obtained mature adipocytes. We further show that these effects are dependent on TGFβ2 but not TGFβ1 signaling: TGFβ2 deficiency decreases adipocyte UCP1 expression, whereas TGFβ2 treatment increases it. The activity of the LTBP3–TGFβ2 axis that we delineate herein also significantly correlates with UCP1 expression in human white adipose tissue (WAT), suggesting an important role in regulating WAT browning as well. Conclusions These results provide evidence that LTBP3, via TGFβ2, plays an important role in promoting brown adipogenesis by modulating UCP1 expression and mitochondrial oxygen consumption. Inhibition of LTBP2 and LTBP3 reduces secretion of TGFβ2. Both knockout of LTBP2/3 or TGFβ2 inhibit UCP1 expression and mitochondrial respiration in human adipocytes. Expression of TGFβ2 correlates with UCP1 expression in human adipose tissue. Treatment with TGFβ2 rescues inhibition of UCP1 by LTBP knockout during adipogenesis.
Collapse
Affiliation(s)
- D Halbgebauer
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany; Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - J Roos
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - J B Funcke
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - H Neubauer
- Cardiometabolic Diseases Research, Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - B S Hamilton
- Cardiometabolic Diseases Research, Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - E Simon
- Global Computational Biology and Digital Sciences, Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - E Z Amri
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | - K M Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - M Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - P Fischer-Posovszky
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - D Tews
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany; Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
150
|
Löffler MC, Betz MJ, Blondin DP, Augustin R, Sharma AK, Tseng YH, Scheele C, Zimdahl H, Mark M, Hennige AM, Wolfrum C, Langhans W, Hamilton BS, Neubauer H. Challenges in tackling energy expenditure as obesity therapy: From preclinical models to clinical application. Mol Metab 2021; 51:101237. [PMID: 33878401 PMCID: PMC8122111 DOI: 10.1016/j.molmet.2021.101237] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/31/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND A chronic imbalance of energy intake and energy expenditure results in excess fat storage. The obesity often caused by this overweight is detrimental to the health of millions of people. Understanding both sides of the energy balance equation and their counter-regulatory mechanisms is critical to the development of effective therapies to treat this epidemic. SCOPE OF REVIEW Behaviors surrounding ingestion have been reviewed extensively. This review focuses more specifically on energy expenditure regarding bodyweight control, with a particular emphasis on the organs and attractive metabolic processes known to reduce bodyweight. Moreover, previous and current attempts at anti-obesity strategies focusing on energy expenditure are highlighted. Precise measurements of energy expenditure, which consist of cellular, animal, and human models, as well as measurements of their translatability, are required to provide the most effective therapies. MAJOR CONCLUSIONS A precise understanding of the components surrounding energy expenditure, including tailored approaches based on genetic, biomarker, or physical characteristics, must be integrated into future anti-obesity treatments. Further comprehensive investigations are required to define suitable treatments, especially because the complex nature of the human perspective remains poorly understood.
Collapse
Affiliation(s)
- Mona C Löffler
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Matthias J Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
| | - Denis P Blondin
- Department of Medicine, Division of Neurology, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, QC, Canada
| | - Robert Augustin
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Anand K Sharma
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Section on Integrative Physiology and Metabolism, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Denmark
| | - Heike Zimdahl
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Michael Mark
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Anita M Hennige
- Therapeutic Area CardioMetabolism & Respiratory, Boehringer Ingelheim International GmbH, Biberach, Germany
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Department of Health Sciences and Technology, ETH Zürich, Switzerland
| | - Bradford S Hamilton
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Heike Neubauer
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany.
| |
Collapse
|