1
|
Tian J, Fan J, Zhang T. Mitochondria as a target for exercise-mitigated type 2 diabetes. J Mol Histol 2023; 54:543-557. [PMID: 37874501 DOI: 10.1007/s10735-023-10158-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 09/17/2023] [Indexed: 10/25/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is one of most common metabolic diseases and continues to be a leading cause of death worldwide. Although great efforts have been made to elucidate the pathogenesis of diabetes, the underlying mechanism still remains unclear. Notably, overwhelming evidence has demonstrated that mitochondria are tightly correlated with the development of T2DM, and the defects of mitochondrial function in peripheral insulin-responsive tissues, such as skeletal muscle, liver and adipose tissue, are crucial drivers of T2DM. Furthermore, exercise training is considered as an effective stimulus for improving insulin sensitivity and hence is regarded as the best strategy to prevent and treat T2DM. Although the precise mechanisms by which exercise alleviates T2DM are not fully understood, mitochondria may be critical for the beneficial effects of exercise.
Collapse
Affiliation(s)
- Jingjing Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China
| | - Jingcheng Fan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China
| | - Tan Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China.
| |
Collapse
|
2
|
Nedergaard J, von Essen G, Cannon B. Brown adipose tissue: can it keep us slim? A discussion of the evidence for and against the existence of diet-induced thermogenesis in mice and men. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220220. [PMID: 37661736 PMCID: PMC10475870 DOI: 10.1098/rstb.2022.0220] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/12/2023] [Indexed: 09/05/2023] Open
Abstract
The issue under discussion here is whether a decrease in the degree of UCP1 activity (and brown adipose tissue activity in general) could be a cause of obesity in humans. This possibility principally requires the existence of the phenomenon of diet-induced thermogenesis. Obesity could be a consequence of a reduced functionality of diet-induced thermogenesis. Experiments in mice indicate that diet-induced thermogenesis exists and is dependent on the presence of UCP1 and thus of brown adipose tissue activity. Accordingly, many (but not all) experiments indicate that in the absence of UCP1, mice become obese. Whether similar mechanisms exist in humans is still unknown. A series of studies have indicated a correlation between obesity and low brown adipose tissue activity, but it may be so that the obesity itself may influence the estimates of brown adipose tissue activity (generally glucose uptake), partly explaining the relationship. Estimates of brown adipose tissue catabolizing activity would seem to indicate that it may possess a capacity sufficient to help maintain body weight, and obesity would thus be aggravated in its absence. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part II)'.
Collapse
Affiliation(s)
- Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Gabriella von Essen
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| |
Collapse
|
3
|
Abstract
Recent advances in pharmacotherapies that promote appetite suppression have shown remarkable weight loss. Therapies targeting energy expenditure lag behind, and as such none have yet been identified to be safe and efficacious for sustaining negative energy balance toward weight loss. Multiple energy dissipating pathways have been identified in adipose tissue and muscle. The molecular effectors of some of these pathways have been identified, but much is still left to be learned about their regulation. Understanding the molecular underpinnings of metabolic inefficiency in adipose tissue and muscle is required if these pathways are to be therapeutically targeted in the context of obesity and obesity-accelerated diseases.
Collapse
Affiliation(s)
- Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
4
|
Martí-Pàmies Í, Thoonen R, Morley M, Graves L, Tamez J, Caplan A, McDaid K, Yao V, Hindle A, Gerszten RE, Laurie A. Farrell, Li L, Tseng YH, Profeta G, Buys ES, Bloch DB, Scherrer-Crosbie M. Brown Adipose Tissue and BMP3b Decrease Injury in Cardiac Ischemia-Reperfusion. Circ Res 2023; 133:353-365. [PMID: 37462036 PMCID: PMC10528340 DOI: 10.1161/circresaha.122.322337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 07/07/2023] [Indexed: 08/05/2023]
Abstract
BACKGROUND Despite advances in treatment, myocardial infarction (MI) is a leading cause of heart failure and death worldwide, with both ischemia and reperfusion (I/R) causing cardiac injury. A previous study using a mouse model of nonreperfused MI showed activation of brown adipose tissue (BAT). Recent studies showed that molecules secreted by BAT target the heart. We investigated whether BAT attenuates cardiac injury in I/R and sought to identify potential cardioprotective proteins secreted by BAT. METHODS Myocardial I/R surgery with or without BAT transplantation was performed in wild-type (WT) mice and in mice with impaired BAT function (uncoupling protein 1 [Ucp1]-deficient mice). To identify potential cardioprotective factors produced by BAT, RNA-seq (RNA sequencing) was performed in BAT from WT and Ucp1-/- mice. Subsequently, myocardial I/R surgery with or without BAT transplantation was performed in Bmp3b (bone morphogenetic protein 3b)-deficient mice, and WT mice subjected to myocardial I/R were treated using BMP3b. RESULTS Dysfunction of BAT in mice was associated with larger MI size after I/R; conversely, augmenting BAT by transplantation decreased MI size. We identified Bmp3b as a protein secreted by BAT after I/R. Compared with WT mice, Bmp3b-deficient mice developed larger MIs. Increasing functional BAT by transplanting BAT from WT mice to Bmp3b-deficient mice reduced I/R injury whereas transplanting BAT from Bmp3b-deficient mice did not. Treatment of WT mice with BMP3b before reperfusion decreased MI size. The cardioprotective effect of BMP3b was mediated through SMAD1/5/8. In humans, the plasma level of BMP3b increased after MI and was positively correlated with the extent of cardiac injury. CONCLUSIONS The results of this study suggest a cardioprotective role of BAT and BMP3b, a protein secreted by BAT, in a model of I/R injury. Interventions increasing BMP3b levels or targeting Smad 1/5 may represent novel therapeutic approaches to decrease myocardial damage in I/R injury.
Collapse
Affiliation(s)
- Íngrid Martí-Pàmies
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Robrecht Thoonen
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, United States
| | - Michael Morley
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Lauren Graves
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Jesus Tamez
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Alex Caplan
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Kendra McDaid
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Vincent Yao
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, United States
| | - Allyson Hindle
- Anesthesia Center for Critical Care Research, Massachusetts General Hospital, Boston, MA, United States
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Laurie A. Farrell
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Li Li
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Yu-Hua Tseng
- 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
| | - Emmanuel S Buys
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, United States
| | - Donald B Bloch
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, United States
- The Center for Immunology and Inflammatory Diseases and the Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, United States
| | - Marielle Scherrer-Crosbie
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
5
|
Jiang T, Su D, Liu X, Wang Y, Wang L. Transcriptomic Analysis Reveals Fibroblast Growth Factor 11 (FGF11) Role in Brown Adipocytes in Thermogenic Regulation of Goats. Int J Mol Sci 2023; 24:10838. [PMID: 37446019 DOI: 10.3390/ijms241310838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/18/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Brown adipose tissue (BAT) is the main site of adaptive thermogenesis, generates heat to maintain body temperature upon cold exposure, and protects against obesity by promoting energy expenditure. RNA-seq analysis revealed that FGF11 is enriched in BAT. However, the functions and regulatory mechanisms of FGF11 in BAT thermogenesis are still limited. In this study, we found that FGF11 was significantly enriched in goat BAT compared with white adipose tissue (WAT). Gain- and loss-of-function experiments revealed that FGF11 promoted differentiation and thermogenesis in brown adipocytes. However, FGF11 had no effect on white adipocyte differentiation. Furthermore, FGF11 promoted the expression of the UCP1 protein and an EBF2 element was responsible for UCP1 promoter activity. Additionally, FGF11 induced UCP1 gene expression through promoting EBF2 binding to the UCP1 promoter. These results revealed that FGF11 promotes differentiation and thermogenesis in brown adipocytes but not in white adipocytes of goats. These findings provide evidence for FGF11 and transcription factor regulatory functions in controlling brown adipose thermogenesis of goats.
Collapse
Affiliation(s)
- Tingting Jiang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Duo Su
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin Liu
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Wang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Linjie Wang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
6
|
Zu Y, Pahlavani M, Ramalingam L, Jayarathne S, Andrade J, Scoggin S, Festuccia WT, Kalupahana NS, Moustaid-Moussa N. Temperature-Dependent Effects of Eicosapentaenoic Acid (EPA) on Browning of Subcutaneous Adipose Tissue in UCP1 Knockout Male Mice. Int J Mol Sci 2023; 24:ijms24108708. [PMID: 37240054 DOI: 10.3390/ijms24108708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Uncoupling protein 1 (UCP1) plays a central role in thermogenic tissues by uncoupling cellular respiration to dissipate energy. Beige adipocytes, an inducible form of thermogenic cells in subcutaneous adipose tissue (SAT), have become a major focus in obesity research. We have previously shown that eicosapentaenoic acid (EPA) ameliorated high-fat diet (HFD)-induced obesity by activating brown fat in C57BL/6J (B6) mice at thermoneutrality (30 °C), independently of UCP1. Here, we investigated whether ambient temperature (22 °C) impacts EPA effects on SAT browning in wild-type (WT) and UCP1 knockout (KO) male mice and dissected underlying mechanisms using a cell model. We observed resistance to diet-induced obesity in UCP1 KO mice fed HFD at ambient temperature, with significantly higher expression of UCP1-independent thermogenic markers, compared to WT mice. These markers included the fibroblast growth factor 21 (FGF21) and sarco/endoplasmic reticulum Ca2+-ATPase 2b (SERCA2b), suggesting the indispensable role of temperature in beige fat reprogramming. Surprisingly, although EPA induced thermogenic effects in SAT-derived adipocytes harvested from both KO and WT mice, EPA only increased thermogenic gene and protein expression in the SAT of UCP1 KO mice housed at ambient temperature. Collectively, our findings indicate that the thermogenic effects of EPA, which are independent of UCP1, occur in a temperature-dependent manner.
Collapse
Affiliation(s)
- Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Mandana Pahlavani
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Shasika Jayarathne
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Jose Andrade
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Shane Scoggin
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - William T Festuccia
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Nishan S Kalupahana
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Department of Physiology, Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
7
|
Hou D, Chen H, Jia T, Zhang L, Gao W, Chen S, Zhu W. Analysis of differential metabolites and metabolic pathways in adipose tissue of tree shrews (Tupaia belangeri) under gradient cooling acclimation. J Therm Biol 2023; 112:103406. [PMID: 36796882 DOI: 10.1016/j.jtherbio.2022.103406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/27/2022] [Accepted: 11/27/2022] [Indexed: 12/13/2022]
Abstract
In order to investigate the influence of gradient cooling acclimation on body mass regulation in tree shrews (Tupaia belangeri), white adipose tissue (WAT) and brown adipose tissue (BAT) in T. belangeri between the control group and gradient cooling acclimation group on day 56 were collected, body mass, food intake, thermogenic capacity, differential metabolites, and related metabolic pathways in WAT and BAT were measured, the changes of differential metabolites were analyzed by non-targeted metabolomics method based on liquid chromatography-mass spectrometry. The results shown that gradient cooling acclimation significantly increased body mass, food intake, resting metabolic rate (RMR), non-shivering thermogenesis (NST), and masses of WAT and BAT. 23 significant differential metabolites in WAT between the gradient cooling acclimation group and the control group, of which the relative contents of 13 differential metabolites were up-regulated and 10 differential metabolites were down-regulated. 27 significant differential metabolites in BAT, of which 18 differential metabolites decreased and 9 differential metabolites increased. 15 differential metabolic pathways in WAT, 8 differential metabolic pathways in BAT, and 4 differential metabolic pathways involved in both WAT and BAT, including Purine metabolism, Pyrimidine metabolism, Glycerol phosphate metabolism, Arginine and proline metabolism, respectively. All of the above results suggested that T. belangeri could use different metabolites of adipose tissue to withstand low temperature environments and enhance their survival.
Collapse
Affiliation(s)
- Dongmin Hou
- Key Laboratory of Ecological Adaptive Evolution and Conservation on Animals-Plants in Southwest Mountain Ecosystem of Yunnan, School of Life Sciences, Yunnan Normal University, Kunming, 650500, China
| | - Huibao Chen
- Key Laboratory of Ecological Adaptive Evolution and Conservation on Animals-Plants in Southwest Mountain Ecosystem of Yunnan, School of Life Sciences, Yunnan Normal University, Kunming, 650500, China
| | - Ting Jia
- Yunnan University of Business Management, Kunming, 650106, China
| | - Lin Zhang
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Wenrong Gao
- School of Biological Resources and Food Engineering, Qujing Normal University, Qujing, 655011, China
| | - Simeng Chen
- Key Laboratory of Ecological Adaptive Evolution and Conservation on Animals-Plants in Southwest Mountain Ecosystem of Yunnan, School of Life Sciences, Yunnan Normal University, Kunming, 650500, China
| | - Wanlong Zhu
- Key Laboratory of Ecological Adaptive Evolution and Conservation on Animals-Plants in Southwest Mountain Ecosystem of Yunnan, School of Life Sciences, Yunnan Normal University, Kunming, 650500, China.
| |
Collapse
|
8
|
Srinivas V, Molangiri A, Varma S, Mallepogu A, Kona SR, Ibrahim A, Duttaroy AK, Basak S. Maternal omega-3 fatty acid deficiency affects fetal thermogenic development and postnatal musculoskeletal growth in mice. J Nutr Biochem 2023; 112:109218. [PMID: 36375730 DOI: 10.1016/j.jnutbio.2022.109218] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
Maternal omega-3 (n-3) polyunsaturated fatty acids (PUFAs) deficiency can affect offspring's adiposity and metabolism by modulating lipid and glucose metabolism. However, the impact of n-3 PUFA deficiency on the development of fetal thermogenesis and its consequences is not reported. Using an n-3 PUFA deficient mice, we assessed fetal interscapular brown adipose tissue (iBAT), body fat composition, insulin growth factor-1 (IGF-1), glucose transporters (GLUTs), and expression of lipid storage & metabolic proteins in the offspring. The n-3 PUFA deficiency did not change the pups' calorie intake, organ weight, and body weight. However, the offspring's skeletal growth was altered due to excess fat to lean mass, reduced tibia & femur elongation, dysregulated IGF-1 in the mother and pups (P< .05). Localization of uncoupling protein 1 (UCP1) in iBAT exhibited a reduced expression in the deficient fetus. Further, UCP1, GLUT1, GPR120 were downregulated while FABP3, ADRP, GLUT4 expressions were upregulated in the BAT of the deficient offspring (P< .05). The deficiency decreased endogenous conversion of the n-3 LCPUFAs from their precursors and upregulated SCD1, FASN, and MFSD2A mRNAs in the liver (P< .05). An altered musculoskeletal growth in the offspring is associated with impaired browning of the fetal adipose, dysregulated thermogenesis, growth hormone, and expression of glucose and fatty acid metabolic mediators due to maternal n-3 PUFA deficiency. BAT had higher metabolic sensitivity compared to WAT in n-3 PUFA deficiency. Maternal n-3 PUFA intake may prevent excess adiposity by modulating fetal development of thermogenesis and skeletal growth dynamics in the mice offspring.
Collapse
Affiliation(s)
- Vilasagaram Srinivas
- National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Archana Molangiri
- National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Saikanth Varma
- National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Aswani Mallepogu
- National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Suryam Reddy Kona
- National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Ahamed Ibrahim
- National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway
| | - Sanjay Basak
- National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India.
| |
Collapse
|
9
|
Choi KM, Cho SH, Kim JH, Kim ARL, Kong X, Yoon JC. CFTR regulates brown adipocyte thermogenesis via the cAMP/PKA signaling pathway. J Cyst Fibros 2023; 22:132-139. [PMID: 36088207 DOI: 10.1016/j.jcf.2022.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 07/12/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) is characterized by reduced growth and lower body weight, which are multifactorial. CF mouse models lack key disease characteristics that predispose to a negative energy balance, such as pulmonary infections or exocrine pancreatic insufficiency, and yet they still exhibit a growth defect and an abnormally increased energy expenditure. Whether adipocyte thermogenesis contributes to the elevated resting energy expenditure in CF mice is unknown. METHODS We examined the expression of CFTR in thermogenic brown adipose tissue (BAT) and investigated a functional role for CFTR using BAT-specific CFTR null mice (CFTRBATKO). RESULTS The CFTR protein is expressed in mouse BAT at levels comparable to those in the lungs. BAT-specific inactivation of CFTR in mice increases whole-body energy expenditure associated with sympathetic stimulation by cold exposure. Weight gain on a high-fat diet is attenuated in these mice. However, CFTR-deficient brown adipocytes themselves have impaired, rather than enhanced, thermogenic responses. These cells feature decreased lipolysis and blunted activation of the cAMP/PKA signaling pathway in response to adrenergic stimulation. This suggests that compensatory heat production in other tissues likely accounts for the increased systemic energy expenditure seen in CFTRBATKO mice. CONCLUSIONS Our data reveal a new role for CFTR in the regulation of adipocyte thermogenesis.
Collapse
Affiliation(s)
- Kyung-Mi Choi
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, California 95616, USA; Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Sung-Hee Cho
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, California 95616, USA
| | - Jung Hak Kim
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, California 95616, USA
| | - Ae-Rhee Lilian Kim
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, California 95616, USA
| | - Xiangmudong Kong
- Department of Surgical and Radiological Sciences, University of California Davis School of Veterinary Medicine, Davis, California 95616, USA
| | - John C Yoon
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, California 95616, USA.
| |
Collapse
|
10
|
Daneshyar S, Ghasemnian A, Mirakhori Z, Daneshyar S. The effect of high fat diet and endurance training on newly discovery of nonshivering-thermogenic factors under thermoneutrality in mice. Sci Sports 2022. [DOI: 10.1016/j.scispo.2022.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
11
|
Čater M, Bombek LK. Protective Role of Mitochondrial Uncoupling Proteins against Age-Related Oxidative Stress in Type 2 Diabetes Mellitus. Antioxidants (Basel) 2022; 11:antiox11081473. [PMID: 36009191 PMCID: PMC9404801 DOI: 10.3390/antiox11081473] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
The accumulation of oxidative damage to DNA and other biomolecules plays an important role in the etiology of aging and age-related diseases such as type 2 diabetes mellitus (T2D), atherosclerosis, and neurodegenerative disorders. Mitochondrial DNA (mtDNA) is especially sensitive to oxidative stress. Mitochondrial dysfunction resulting from the accumulation of mtDNA damage impairs normal cellular function and leads to a bioenergetic crisis that accelerates aging and associated diseases. Age-related mitochondrial dysfunction decreases ATP production, which directly affects insulin secretion by pancreatic beta cells and triggers the gradual development of the chronic metabolic dysfunction that characterizes T2D. At the same time, decreased glucose oxidation in skeletal muscle due to mitochondrial damage leads to prolonged postprandial blood glucose rise, which further worsens glucose homeostasis. ROS are not only highly reactive by-products of mitochondrial respiration capable of oxidizing DNA, proteins, and lipids but can also function as signaling and effector molecules in cell membranes mediating signal transduction and inflammation. Mitochondrial uncoupling proteins (UCPs) located in the inner mitochondrial membrane of various tissues can be activated by ROS to protect cells from mitochondrial damage. Mitochondrial UCPs facilitate the reflux of protons from the mitochondrial intermembrane space into the matrix, thereby dissipating the proton gradient required for oxidative phosphorylation. There are five known isoforms (UCP1-UCP5) of mitochondrial UCPs. UCP1 can indirectly reduce ROS formation by increasing glutathione levels, thermogenesis, and energy expenditure. In contrast, UCP2 and UCP3 regulate fatty acid metabolism and insulin secretion by beta cells and modulate insulin sensitivity. Understanding the functions of UCPs may play a critical role in developing pharmacological strategies to combat T2D. This review summarizes the current knowledge on the protective role of various UCP homologs against age-related oxidative stress in T2D.
Collapse
Affiliation(s)
- Maša Čater
- Correspondence: (M.Č.); (L.K.B.); Tel.: +386-2-2345-847 (L.K.B.)
| | | |
Collapse
|
12
|
Chen H, Zhao F, Chen K, Guo Y, Liang Y, Zhao H, Chen S. Exposure of zebrafish to a cold environment triggered cellular autophagy in zebrafish liver. JOURNAL OF FISH DISEASES 2022; 45:991-1000. [PMID: 35395109 DOI: 10.1111/jfd.13620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 06/14/2023]
Abstract
Water temperature is the major ecophysiological factor for fish survival in nature and aquaculture. Compared with many homeotherms, fish can survive prolonged periods under the condition of low temperature. However, the metabolic strategies of the liver under a cold environment are still unknown in this species. In our present study, adult zebrafish were exposed to a cold or cold plus starvation environment to analyse the morphological characteristics of hepatocytes by light microscopy and transmission electron microscopy (TEM). The fish livers were dissected and observed under a microscope, and the liver size and shape appeared normal in all groups. Periodic acid-Schiff and TEM analysis showed that hepatic glycogen was significantly lower in zebrafish exposed to cold acclimation (CF group) than that zebrafish at the control water temperature (CT group). Moreover, qPCR and IHC results indicated that the expression of PYGL (a key enzyme involved in glycogenolysis) markedly increased in the CF group. After cold plus starvation treatment (CS group), autophagy activity was significantly enhanced and numerous mitophagic vacuoles were present in the cytoplasm of hepatocytes. In conclusion, hepatic glycogen was first mobilizing to supply energy, and then autophagy, especially mitophagy, played vital roles during nutrient deprivation in fish species.
Collapse
Affiliation(s)
- Hong Chen
- College of Veterinary Medicine, Northwest A&F University, Shaanxi Province, Yangling, China
| | - Fange Zhao
- College of Veterinary Medicine, Northwest A&F University, Shaanxi Province, Yangling, China
| | - Kexing Chen
- College of Veterinary Medicine, Northwest A&F University, Shaanxi Province, Yangling, China
| | - Yihan Guo
- College of Veterinary Medicine, Northwest A&F University, Shaanxi Province, Yangling, China
| | - Yue Liang
- College of Veterinary Medicine, Northwest A&F University, Shaanxi Province, Yangling, China
| | - Huiying Zhao
- College of Veterinary Medicine, Northwest A&F University, Shaanxi Province, Yangling, China
| | - Shulin Chen
- College of Veterinary Medicine, Northwest A&F University, Shaanxi Province, Yangling, China
| |
Collapse
|
13
|
Huang Z, Zhang Z, Moazzami Z, Heck R, Hu P, Nanda H, Ren K, Sun Z, Bartolomucci A, Gao Y, Chung D, Zhu W, Shen S, Ruan HB. Brown adipose tissue involution associated with progressive restriction in progenitor competence. Cell Rep 2022; 39:110575. [PMID: 35417710 DOI: 10.1016/j.celrep.2022.110575] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/10/2022] [Accepted: 03/04/2022] [Indexed: 11/03/2022] Open
Abstract
Human brown adipose tissue (BAT) undergoes progressive involution. This involution process is not recapitulated in rodents, and the underlying mechanisms are poorly understood. Here we show that the interscapular BAT (iBAT) of rabbits whitens rapidly during early adulthood. The transcriptomic remodeling and identity switch of mature adipocytes are accompanied by loss of brown adipogenic competence of progenitors. Single-cell RNA sequencing reveals that rabbit and human iBAT progenitors highly express the FSTL1 gene. When iBAT involutes in rabbits, adipocyte progenitors reduce FSTL1 expression and are refractory to brown adipogenic recruitment. Conversely, FSTL1 is constitutively expressed in mouse iBAT to sustain WNT signaling and prevent involution. Progenitor incompetence and iBAT paucity can be induced in mice by genetic deletion of the Fstl1 gene or ablation of Fstl1+ progenitors. Our results highlight the hierarchy and dynamics of the BAT progenitor compartment and implicate the functional incompetence of FSTL1-expressing progenitors in BAT involution.
Collapse
Affiliation(s)
- Zan Huang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China; Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| | - Zengdi Zhang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Zahra Moazzami
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Ryan Heck
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Ping Hu
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, China
| | - Hezkiel Nanda
- Institute for Health Informatics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Kaiqun Ren
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; College of Medicine, Hunan Normal University, Changsha, Hunan 410081, China
| | - Zequn Sun
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Yan Gao
- Department of Human Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Steven Shen
- Institute for Health Informatics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Clinical Translational Science Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
14
|
Oeckl J, Janovska P, Adamcova K, Bardova K, Brunner S, Dieckmann S, Ecker J, Fromme T, Funda J, Gantert T, Giansanti P, Hidrobo MS, Kuda O, Kuster B, Li Y, Pohl R, Schmitt S, Schweizer S, Zischka H, Zouhar P, Kopecky J, Klingenspor M. Loss of UCP1 function augments recruitment of futile lipid cycling for thermogenesis in murine brown fat. Mol Metab 2022; 61:101499. [PMID: 35470094 PMCID: PMC9097615 DOI: 10.1016/j.molmet.2022.101499] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/12/2022] [Accepted: 04/12/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Josef Oeckl
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Petra Janovska
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Czech Republic
| | - Katerina Adamcova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Czech Republic
| | - Kristina Bardova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Czech Republic
| | - Sarah Brunner
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Sebastian Dieckmann
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Josef Ecker
- ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Tobias Fromme
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Jiri Funda
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Czech Republic
| | - Thomas Gantert
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Piero Giansanti
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; Bavarian Center for Biomolecular Mass Spectrometry, Technical University of Munich, Freising, Germany
| | - Maria Soledad Hidrobo
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Ondrej Kuda
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Czech Republic
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; Bavarian Center for Biomolecular Mass Spectrometry, Technical University of Munich, Freising, Germany
| | - Yongguo Li
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Radek Pohl
- NMR spectroscopy, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Czech Republic
| | - Sabine Schmitt
- Institute of Toxicology and Environmental Hygiene, School of Medicine, Technical University of Munich, Munich, Germany
| | - Sabine Schweizer
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Hans Zischka
- Institute of Toxicology and Environmental Hygiene, School of Medicine, Technical University of Munich, Munich, Germany; Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, Munich, Germany
| | - Petr Zouhar
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Czech Republic
| | - Jan Kopecky
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Czech Republic.
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany.
| |
Collapse
|
15
|
Li Y, Fromme T. Uncoupling Protein 1 Does Not Produce Heat without Activation. Int J Mol Sci 2022; 23:2406. [PMID: 35269549 PMCID: PMC8910648 DOI: 10.3390/ijms23052406] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial uncoupling protein 1 (UCP1) is the crucial mechanistic component of heat production in classical brown fat and the newly identified beige or brite fat. Thermogenesis inevitably comes at a high energetic cost and brown fat, ultimately, is an energy-wasting organ. A constrained strategy that minimizes brown fat activity unless obligate will have been favored during natural selection to safeguard metabolic thriftiness. Accordingly, UCP1 is constitutively inhibited and is inherently not leaky without activation. It follows that increasing brown adipocyte number or UCP1 abundance genetically or pharmacologically does not lead to an automatic increase in thermogenesis or subsequent metabolic consequences in the absence of a plausible route of concomitant activation. Despite its apparent obviousness, this tenet is frequently ignored. Consequently, incorrect conclusions are often drawn from increased BAT or brite/beige depot mass, e.g., predicting or causally linking beneficial metabolic effects. Here, we highlight the inherently inactive nature of UCP1, with a particular emphasis on the molecular brakes and releases of UCP1 activation under physiological conditions. These controls of UCP1 activity represent potential targets of therapeutic interventions to unlock constraints and efficiently harness the energy-expending potential of brown fat to prevent and treat obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Yongguo Li
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Tobias Fromme
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| |
Collapse
|
16
|
Dieckmann S, Strohmeyer A, Willershäuser M, Maurer SF, Wurst W, Marschall S, de Angelis MH, Kühn R, Worthmann A, Fuh MM, Heeren J, Köhler N, Pauling JK, Klingenspor M. Susceptibility to diet-induced obesity at thermoneutral conditions is independent of UCP1. Am J Physiol Endocrinol Metab 2022; 322:E85-E100. [PMID: 34927460 DOI: 10.1152/ajpendo.00278.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Activation of uncoupling protein 1 (UCP1) in brown adipose tissue (BAT) upon cold stimulation leads to substantial increase in energy expenditure to defend body temperature. Increases in energy expenditure after a high-caloric food intake, termed diet-induced thermogenesis, are also attributed to BAT. These properties render BAT a potential target to combat diet-induced obesity. However, studies investigating the role of UCP1 to protect against diet-induced obesity are controversial and rely on the phenotyping of a single constitutive UCP1-knockout model. To address this issue, we generated a novel UCP1-knockout model by Cre-mediated deletion of exon 2 in the UCP1 gene. We studied the effect of constitutive UCP1 knockout on metabolism and the development of diet-induced obesity. UCP1 knockout and wild-type mice were housed at 30°C and fed a control diet for 4 wk followed by 8 wk of high-fat diet. Body weight and food intake were monitored continuously over the course of the study, and indirect calorimetry was used to determine energy expenditure during both feeding periods. Based on Western blot analysis, thermal imaging and noradrenaline test, we confirmed the lack of functional UCP1 in knockout mice. However, body weight gain, food intake, and energy expenditure were not affected by loss of UCP1 function during both feeding periods. We introduce a novel UCP1-KO mouse enabling the generation of conditional UCP1-knockout mice to scrutinize the contribution of UCP1 to energy metabolism in different cell types or life stages. Our results demonstrate that UCP1 does not protect against diet-induced obesity at thermoneutrality.NEW & NOTEWORTHY We provide evidence that the abundance of UCP1 does not influence energy metabolism at thermoneutrality studying a novel Cre-mediated UCP1-KO mouse model. This model will be a foundation for a better understanding of the contribution of UCP1 in different cell types or life stages to energy metabolism.
Collapse
Affiliation(s)
- Sebastian Dieckmann
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Akim Strohmeyer
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Monja Willershäuser
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Stefanie F Maurer
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Germany
- TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Munich, Germany
| | - Susan Marschall
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Martin Hrabe de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ralf Kühn
- Institute of Developmental Genetics, Helmholtz Zentrum München, Germany
| | - Anna Worthmann
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marceline M Fuh
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nikolai Köhler
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Josch K Pauling
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| |
Collapse
|
17
|
Yu H, Nie C, Zhou Y, Wang X, Wang H, Shi X. Tolerance to Glucose and Lipid High Metabolic Reactions After Burns in an Obese Rat Model. J Burn Care Res 2022; 43:1-8. [PMID: 34520555 DOI: 10.1093/jbcr/irab163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The goal of this study was to determine what effect obese body weight and a burn injury can have on the metabolism of glucose and lipids in rats. We used a 3 * 3 factorial model design to provide basic glucose and lipid metabolic data characterizing the interaction between different weight and burn injury groups. Two hundred Sprague Dawley rats were categorized into three weight groups (normal, overweight, obese) and then further divided into control, second-degree, and third-degree burn groups. Our model compared interactions between weight and burn injury factors according to the above groups. Blood glucose and lipid metabolism indicators were monitored on the 1st, 3rd, 7th, and 14th days after burn injury occurred, and burned skin and blood samples were collected for testing. Compared with the normal weight group, the overweight group's fast blood glucose, fast insulin, and homeostasis model assessment of insulin resistance were higher (P < .05), and FBG in the obese group was higher than the normal weight group (P < .05). Burn injuries combined with obese body weight had an interactive effect on fast blood glucose, fast insulin, and homeostasis model assessment of insulin resistance after burn injury (P < .05). Burn injury combined with obese body weight had an interaction on low-density lipoprotein cholesterol on the 3rd day after burn injury (P < .05). Burn injury combined with obese weight had no interaction on triglyceride, total cholesterol, and high-density lipoprotein cholesterol (P > .05). Rats in the overweight and obese weight groups were observed to develop an adaptation and tolerance to a higher metabolic rate after burn injuries occurred.
Collapse
Affiliation(s)
- Huiting Yu
- Department of Epidemiology and Health Statistics, School of Public Health, Zunyi Medical University, Guizhou, China
- Hospital Infection Control Department, Qiaokou District Maternal and Child Health Hospital, Wuhan, Hubei, China
| | - Chan Nie
- Department of Epidemiology and Health Statistics, School of Public Health, Zunyi Medical University, Guizhou, China
- Department of Epidemiology, Guiyang Center for Disease Control and Prevention, Guiyang, Guizhou, China
| | - Yanna Zhou
- Department of Epidemiology and Health Statistics, School of Public Health, Zunyi Medical University, Guizhou, China
| | - Xue Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Zunyi Medical University, Guizhou, China
| | - Haiyan Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Zunyi Medical University, Guizhou, China
| | - Xiuquan Shi
- Department of Epidemiology and Health Statistics, School of Public Health, Zunyi Medical University, Guizhou, China
| |
Collapse
|
18
|
Romanelli SM, Lewis KT, Nishii A, Rupp AC, Li Z, Mori H, Schill RL, Learman BS, Rhodes CJ, MacDougald OA. BAd-CRISPR: Inducible gene knockout in interscapular brown adipose tissue of adult mice. J Biol Chem 2021; 297:101402. [PMID: 34774798 PMCID: PMC8661024 DOI: 10.1016/j.jbc.2021.101402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/26/2021] [Accepted: 11/09/2021] [Indexed: 12/26/2022] Open
Abstract
CRISPR/Cas9 has enabled inducible gene knockout in numerous tissues; however, its use has not been reported in brown adipose tissue (BAT). Here, we developed the brown adipocyte CRISPR (BAd-CRISPR) methodology to rapidly interrogate the function of one or multiple genes. With BAd-CRISPR, an adeno-associated virus (AAV8) expressing a single guide RNA (sgRNA) is administered directly to BAT of mice expressing Cas9 in brown adipocytes. We show that the local administration of AAV8-sgRNA to interscapular BAT of adult mice robustly transduced brown adipocytes and ablated expression of adiponectin, adipose triglyceride lipase, fatty acid synthase, perilipin 1, or stearoyl-CoA desaturase 1 by >90%. Administration of multiple AAV8 sgRNAs led to simultaneous knockout of up to three genes. BAd-CRISPR induced frameshift mutations and suppressed target gene mRNA expression but did not lead to substantial accumulation of off-target mutations in BAT. We used BAd-CRISPR to create an inducible uncoupling protein 1 (Ucp1) knockout mouse to assess the effects of UCP1 loss on adaptive thermogenesis in adult mice. Inducible Ucp1 knockout did not alter core body temperature; however, BAd-CRISPR Ucp1 mice had elevated circulating concentrations of fibroblast growth factor 21 and changes in BAT gene expression consistent with heat production through increased peroxisomal lipid oxidation. Other molecular adaptations predict additional cellular inefficiencies with an increase in both protein synthesis and turnover, and mitochondria with reduced reliance on mitochondrial-encoded gene expression and increased expression of nuclear-encoded mitochondrial genes. These data suggest that BAd-CRISPR is an efficient tool to speed discoveries in adipose tissue biology.
Collapse
Affiliation(s)
- Steven M Romanelli
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kenneth T Lewis
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Akira Nishii
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Alan C Rupp
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ziru Li
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Hiroyuki Mori
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Rebecca L Schill
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Brian S Learman
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Christopher J Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
19
|
Brandão BB, Poojari A, Rabiee A. Thermogenic Fat: Development, Physiological Function, and Therapeutic Potential. Int J Mol Sci 2021; 22:5906. [PMID: 34072788 PMCID: PMC8198523 DOI: 10.3390/ijms22115906] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
The concerning worldwide increase of obesity and chronic metabolic diseases, such as T2D, dyslipidemia, and cardiovascular disease, motivates further investigations into preventive and alternative therapeutic approaches. Over the past decade, there has been growing evidence that the formation and activation of thermogenic adipocytes (brown and beige) may serve as therapy to treat obesity and its associated diseases owing to its capacity to increase energy expenditure and to modulate circulating lipids and glucose levels. Thus, understanding the molecular mechanism of brown and beige adipocytes formation and activation will facilitate the development of strategies to combat metabolic disorders. Here, we provide a comprehensive overview of pathways and players involved in the development of brown and beige fat, as well as the role of thermogenic adipocytes in energy homeostasis and metabolism. Furthermore, we discuss the alterations in brown and beige adipose tissue function during obesity and explore the therapeutic potential of thermogenic activation to treat metabolic syndrome.
Collapse
Affiliation(s)
- Bruna B. Brandão
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Ankita Poojari
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| | - Atefeh Rabiee
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| |
Collapse
|
20
|
Wang H, Willershäuser M, Li Y, Fromme T, Schnabl K, Bast-Habersbrunner A, Ramisch S, Mocek S, Klingenspor M. Uncoupling protein-1 expression does not protect mice from diet-induced obesity. Am J Physiol Endocrinol Metab 2021; 320:E333-E345. [PMID: 33252252 PMCID: PMC8260371 DOI: 10.1152/ajpendo.00285.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We studied the metabolic phenotype of a novel Ucp1-LUC-iRFP713 knock-in reporter gene mouse model originally generated to monitor endogenous Ucp1 gene expression. Both reporter mice and reporter cells reliably reflected Ucp1 gene expression in vivo and in vitro. We here report an unexpected reduction in UCP1 content in homozygous knock-in (KI) reporter mice. As a result, the thermogenic capacity of KI mice stimulated by norepinephrine was largely blunted, making them more sensitive to an acute cold exposure. In return, these reporter mice with reduced UCP1 expression enabled us to investigate the physiological role of UCP1 in the prevention of weight gain. We observed no substantial differences in body mass across the three genotypes, irrespective of the type of diet or the ambient temperature, possibly due to the insufficient UCP1 activation. Indeed, activation of UCP1 by daily injection of the selective β3-adrenergic receptor agonist CL316,243 resulted in significantly greater reduction of body weight in wild-type mice than in KI mice. Taken together, we conclude that the intact expression of UCP1 is essential for cold-induced thermogenesis but the presence of UCP1 per se does not protect mice from diet-induced obesity.NEW & NOTEWORTHY To study the functional role of UCP1-dependent brown adipose tissue thermogenesis for energy balance, new animal models are needed. By metabolic phenotyping of a novel mouse model with low UCP1 levels in brown fat, we demonstrate that the susceptibility to diet-induced obesity is not increased despite impaired cold-induced thermogenic capacity. Brown fat requires pharmacological activation to promote negative energy balance in diet-induced obese mice.
Collapse
Affiliation(s)
- Hui Wang
- Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Monja Willershäuser
- Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Yongguo Li
- Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Tobias Fromme
- Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Katharina Schnabl
- Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Andrea Bast-Habersbrunner
- Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Samira Ramisch
- Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Sabine Mocek
- Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Martin Klingenspor
- Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany
| |
Collapse
|
21
|
Maurer SF, Dieckmann S, Lund J, Fromme T, Hess AL, Colson C, Kjølbaek L, Astrup A, Gillum MP, Larsen LH, Liebisch G, Amri EZ, Klingenspor M. No Effect of Dietary Fish Oil Supplementation on the Recruitment of Brown and Brite Adipocytes in Mice or Humans under Thermoneutral Conditions. Mol Nutr Food Res 2021; 65:e2000681. [PMID: 33274552 DOI: 10.1002/mnfr.202000681] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/09/2020] [Indexed: 01/06/2023]
Abstract
SCOPE Brown and brite adipocytes within the mammalian adipose organ provide non-shivering thermogenesis and thus, have an exceptional capacity to dissipate chemical energy as heat. Polyunsaturated fatty acids (PUFA) of the n3-series, abundant in fish oil, have been repeatedly demonstrated to enhance the recruitment of thermogenic capacity in these cells, consequently affecting body adiposity and glucose tolerance. These effects are scrutinized in mice housed in a thermoneutral environment and in a human dietary intervention trial. METHODS AND RESULTS Mice are housed in a thermoneutral environment eliminating the superimposing effect of mild cold-exposure on thermogenic adipocyte recruitment. Dietary fish oil supplementation in two different inbred mouse strains neither affects body mass trajectory nor enhances the recruitment of brown and brite adipocytes, both in the presence and absence of a β3-adrenoreceptor agonist imitating the effect of cold-exposure on adipocytes. In line with these findings, dietary fish oil supplementation of persons with overweight or obesity fails to recruit thermogenic adipocytes in subcutaneous adipose tissue. CONCLUSION Thus, the authors' data question the hypothesized potential of n3-PUFA as modulators of adipocyte-based thermogenesis and energy balance regulation.
Collapse
Affiliation(s)
- Stefanie F Maurer
- Chair for Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences, Freising, 85354, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, 85354, Germany
| | - Sebastian Dieckmann
- Chair for Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences, Freising, 85354, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, 85354, Germany
- ZIEL - Institute for Food and Health, Technical University of Munich, Freising, 85354, Germany
| | - Jens Lund
- Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Frederiksberg, DK-1958, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Tobias Fromme
- Chair for Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences, Freising, 85354, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, 85354, Germany
- ZIEL - Institute for Food and Health, Technical University of Munich, Freising, 85354, Germany
| | - Anne Lundby Hess
- Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Frederiksberg, DK-1958, Denmark
| | - Cécilia Colson
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, 06107, France
| | - Louise Kjølbaek
- Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Frederiksberg, DK-1958, Denmark
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Frederiksberg, DK-1958, Denmark
| | - Matthew Paul Gillum
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Lesli Hingstrup Larsen
- Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Frederiksberg, DK-1958, Denmark
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, Regensburg, 93053, Germany
| | - Ez-Zoubir Amri
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, 06107, France
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences, Freising, 85354, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, 85354, Germany
- ZIEL - Institute for Food and Health, Technical University of Munich, Freising, 85354, Germany
| |
Collapse
|
22
|
Kaikaew K, Grefhorst A, Visser JA. Sex Differences in Brown Adipose Tissue Function: Sex Hormones, Glucocorticoids, and Their Crosstalk. Front Endocrinol (Lausanne) 2021; 12:652444. [PMID: 33927694 PMCID: PMC8078866 DOI: 10.3389/fendo.2021.652444] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022] Open
Abstract
Excessive fat accumulation in the body causes overweight and obesity. To date, research has confirmed that there are two types of adipose tissue with opposing functions: lipid-storing white adipose tissue (WAT) and lipid-burning brown adipose tissue (BAT). After the rediscovery of the presence of metabolically active BAT in adults, BAT has received increasing attention especially since activation of BAT is considered a promising way to combat obesity and associated comorbidities. It has become clear that energy homeostasis differs between the sexes, which has a significant impact on the development of pathological conditions such as type 2 diabetes. Sex differences in BAT activity may contribute to this and, therefore, it is important to address the underlying mechanisms that contribute to sex differences in BAT activity. In this review, we discuss the role of sex hormones in the regulation of BAT activity under physiological and some pathological conditions. Given the increasing number of studies suggesting a crosstalk between sex hormones and the hypothalamic-pituitary-adrenal axis in metabolism, we also discuss this crosstalk in relation to sex differences in BAT activity.
Collapse
Affiliation(s)
- Kasiphak Kaikaew
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Aldo Grefhorst
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
| | - Jenny A. Visser
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- *Correspondence: Jenny A. Visser,
| |
Collapse
|
23
|
Role of adiposopathy and physical activity in cardio-metabolic disorder diseases. Clin Chim Acta 2020; 511:243-247. [PMID: 33148528 DOI: 10.1016/j.cca.2020.10.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 01/27/2023]
Abstract
Positive calorie balance disrupts the function of visceral adipose tissue, including the cardiac adipose tissue and the perivascular adipose tissue. The inflammatory and hormonal factors, which are released from adipose tissue, play a central role in inter-organ cross talk, affecting the development of obesity. Excess fat in visceral adipocytes impairs endocrine as well as immune response, leading to multiple aberrant status and posing serious risks to the future health of humans. As confirmed in previous studies, up-regulated pro-inflammatory and down-regulated anti-inflammatory cytokines disturb the communication among muscle, liver, and vasculature. In other words, adiposopathy promote cardio-metabolic risk factors, such as atherosclerosis, hypertension, insulin resistance, dyslipidemia, and pro-thrombotic state, which in turn directly and indirectly promote cardio-metabolic disorder diseases. Increasing evidence from human and animal studies has shown that physical activity restores the size of adipocytes and helps in re-browning of white adipose tissue (WAT). This review summarizes the current evidence on the roles of adiposopathy on cardio-metabolic disorder diseases and the importance of physical activity in restoring the function of adipocytes.
Collapse
|
24
|
Dehvari N, Sato M, Bokhari MH, Kalinovich A, Ham S, Gao J, Nguyen HTM, Whiting L, Mukaida S, Merlin J, Chia LY, Wootten D, Summers RJ, Evans BA, Bengtsson T, Hutchinson DS. The metabolic effects of mirabegron are mediated primarily by β 3 -adrenoceptors. Pharmacol Res Perspect 2020; 8:e00643. [PMID: 32813332 PMCID: PMC7437350 DOI: 10.1002/prp2.643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 12/19/2022] Open
Abstract
The β3 -adrenoceptor agonist mirabegron is approved for use for overactive bladder and has been purported to be useful in the treatment of obesity-related metabolic diseases in humans, including those involving disturbances of glucose homeostasis. We investigated the effect of mirabegron on glucose homeostasis with in vitro and in vivo models, focusing on its selectivity at β-adrenoceptors, ability to cause browning of white adipocytes, and the role of UCP1 in glucose homeostasis. In mouse brown, white, and brite adipocytes, mirabegron-mediated effects were examined on cyclic AMP, UCP1 mRNA, [3 H]-2-deoxyglucose uptake, cellular glycolysis, and O2 consumption. Mirabegron increased cyclic AMP levels, UCP1 mRNA content, glucose uptake, and cellular glycolysis in brown adipocytes, and these effects were either absent or reduced in white adipocytes. In brite adipocytes, mirabegron increased cyclic AMP levels and UCP1 mRNA content resulting in increased UCP1-mediated oxygen consumption, glucose uptake, and cellular glycolysis. The metabolic effects of mirabegron in both brown and brite adipocytes were primarily due to actions at β3 -adrenoceptors as they were largely absent in adipocytes derived from β3 -adrenoceptor knockout mice. In vivo, mirabegron increased whole body oxygen consumption, glucose uptake into brown and inguinal white adipose tissue, and improved glucose tolerance, all effects that required the presence of the β3 -adrenoceptor. Furthermore, in UCP1 knockout mice, the effects of mirabegron on glucose tolerance were attenuated. Thus, mirabegron had effects on cellular metabolism in adipocytes that improved glucose handling in vivo, and were primarily due to actions at the β3 -adrenoceptor.
Collapse
Affiliation(s)
- Nodi Dehvari
- Department of Molecular BiosciencesThe Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Masaaki Sato
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| | - Muhammad Hamza Bokhari
- Department of Molecular BiosciencesThe Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Anastasia Kalinovich
- Department of Molecular BiosciencesThe Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Seungmin Ham
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| | - Jie Gao
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| | - Huong T. M. Nguyen
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| | - Lynda Whiting
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| | - Saori Mukaida
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| | - Jon Merlin
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| | - Ling Yeong Chia
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| | - Denise Wootten
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| | - Roger J. Summers
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| | - Bronwyn A. Evans
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| | - Tore Bengtsson
- Department of Molecular BiosciencesThe Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Dana S. Hutchinson
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVic.Australia
| |
Collapse
|
25
|
Ruan HB. Developmental and functional heterogeneity of thermogenic adipose tissue. J Mol Cell Biol 2020; 12:775-784. [PMID: 32569352 PMCID: PMC7816678 DOI: 10.1093/jmcb/mjaa029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/11/2020] [Accepted: 06/12/2020] [Indexed: 12/18/2022] Open
Abstract
The obesity epidemic continues to rise as a global health challenge. Thermogenic brown and beige adipocytes dissipate chemical energy as heat, providing an opportunity for developing new therapeutics for obesity and related metabolic diseases. Anatomically, brown adipose tissue is distributed as discrete depots, while beige adipocytes exist within certain depots of white adipose tissue. Developmentally, brown and beige adipocytes arise from multiple embryonic progenitor populations that are distinct and overlapping. Functionally, they respond to a plethora of stimuli to engage uncoupling protein 1-dependent and independent thermogenic programs, thus improving systemic glucose homeostasis, lipid metabolism, and the clearance of branched-chain amino acids. In this review, we highlight recent advances in our understanding of the molecular and cellular mechanisms that contribute to the developmental and functional heterogeneity of thermogenic adipose tissue.
Collapse
Affiliation(s)
- Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
26
|
Fischer AW, Behrens J, Sass F, Schlein C, Heine M, Pertzborn P, Scheja L, Heeren J. Brown adipose tissue lipoprotein and glucose disposal is not determined by thermogenesis in uncoupling protein 1-deficient mice. J Lipid Res 2020; 61:1377-1389. [PMID: 32769145 DOI: 10.1194/jlr.ra119000455] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Adaptive thermogenesis is highly dependent on uncoupling protein 1 (UCP1), a protein expressed by thermogenic adipocytes present in brown adipose tissue (BAT) and white adipose tissue (WAT). Thermogenic capacity of human and mouse BAT can be measured by positron emission tomography-computed tomography quantifying the uptake of 18F-fluodeoxyglucose or lipid tracers. BAT activation is typically studied in response to cold exposure or treatment with β-3-adrenergic receptor agonists such as CL316,243 (CL). Currently, it is unknown whether cold-stimulated uptake of glucose or lipid tracers is a good surrogate marker of UCP1-mediated thermogenesis. In metabolic studies using radiolabeled tracers, we found that glucose uptake is increased in mildly cold-activated BAT of Ucp1 -/- versus WT mice kept at subthermoneutral temperature. Conversely, lower glucose disposal was detected after full thermogenic activation achieved by sustained cold exposure or CL treatment. In contrast, uptake of lipoprotein-derived fatty acids into chronically activated thermogenic adipose tissues was substantially increased in UCP1-deficient mice. This effect is linked to higher sympathetic tone in adipose tissues of Ucp1 -/- mice, as indicated by elevated levels of thermogenic genes in BAT and WAT. Thus, glucose and lipoprotein handling does not necessarily reflect UCP1-dependent thermogenic activity, but especially lipid uptake rather mirrors sympathetic activation of adipose tissues.
Collapse
Affiliation(s)
- Alexander W Fischer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Janina Behrens
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frederike Sass
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Schlein
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Pertzborn
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
27
|
Abstract
Perturbations in metabolic processes are associated with diseases such as obesity, type 2 diabetes mellitus, certain infections and some cancers. A resurgence of interest in creatine biology is developing, with new insights into a diverse set of regulatory functions for creatine. This resurgence is primarily driven by technological advances in genetic engineering and metabolism as well as by the realization that this metabolite has key roles in cells beyond the muscle and brain. Herein, we highlight the latest advances in creatine biology in tissues and cell types that have historically received little attention in the field. In adipose tissue, creatine controls thermogenic respiration and loss of this metabolite impairs whole-body energy expenditure, leading to obesity. We also cover the various roles that creatine metabolism has in cancer cell survival and the function of the immune system. Renewed interest in this area has begun to showcase the therapeutic potential that lies in understanding how changes in creatine metabolism lead to metabolic disease.
Collapse
Affiliation(s)
- Lawrence Kazak
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.
- Department of Biochemistry, McGill University, Montreal, QC, Canada.
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
28
|
UCP1-independent thermogenesis. Biochem J 2020; 477:709-725. [PMID: 32059055 DOI: 10.1042/bcj20190463] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/24/2022]
Abstract
Obesity results from energy imbalance, when energy intake exceeds energy expenditure. Brown adipose tissue (BAT) drives non-shivering thermogenesis which represents a powerful mechanism of enhancing the energy expenditure side of the energy balance equation. The best understood thermogenic system in BAT that evolved to protect the body from hypothermia is based on the uncoupling of protonmotive force from oxidative phosphorylation through the actions of uncoupling protein 1 (UCP1), a key regulator of cold-mediated thermogenesis. Similarly, energy expenditure is triggered in response to caloric excess, and animals with reduced thermogenic fat function can succumb to diet-induced obesity. Thus, it was surprising when inactivation of Ucp1 did not potentiate diet-induced obesity. In recent years, it has become clear that multiple thermogenic mechanisms exist, based on ATP sinks centered on creatine, lipid, or calcium cycling, along with Fatty acid-mediated UCP1-independent leak pathways driven by the ADP/ATP carrier (AAC). With a key difference between cold- and diet-induced thermogenesis being the dynamic changes in purine nucleotide (primarily ATP) levels, ATP-dependent thermogenic pathways may play a key role in diet-induced thermogenesis. Additionally, the ubiquitous expression of AAC may facilitate increased energy expenditure in many cell types, in the face of over feeding. Interest in UCP1-independent energy expenditure has begun to showcase the therapeutic potential that lies in refining our understanding of the diversity of biochemical pathways controlling thermogenic respiration.
Collapse
|
29
|
Maurer S, Harms M, Boucher J. The colorful versatility of adipocytes: white-to-brown transdifferentiation and its therapeutic potential in humans. FEBS J 2020; 288:3628-3646. [PMID: 32621398 DOI: 10.1111/febs.15470] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/17/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
Brown and brite adipocytes contribute to energy expenditure through nonshivering thermogenesis. Though these cell types are thought to arise primarily from the de novo differentiation of precursor cells, their abundance is also controlled through the transdifferentiation of mature white adipocytes. Here, we review recent advances in our understanding of the regulation of white-to-brown transdifferentiation, as well as the conversion of brown and brite adipocytes to dormant, white-like fat cells. Converting mature white adipocytes into brite cells or reactivating dormant brown and brite adipocytes has emerged as a strategy to ameliorate human metabolic disorders. We analyze the evidence of learning from mice and how they translate to humans to ultimately scrutinize the relevance of this concept. Moreover, we estimate that converting a small percentage of existing white fat mass in obese subjects into active brite adipocytes could be sufficient to achieve meaningful benefits in metabolism. In conclusion, novel browning agents have to be identified before adipocyte transdifferentiation can be realized as a safe and efficacious therapy.
Collapse
Affiliation(s)
- Stefanie Maurer
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Matthew Harms
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jeremie Boucher
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Lundberg Laboratory for Diabetes Research, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
30
|
Gildner TE, Levy SB. Intersecting vulnerabilities in human biology: Synergistic interactions between climate change and increasing obesity rates. Am J Hum Biol 2020; 33:e23460. [PMID: 32618027 DOI: 10.1002/ajhb.23460] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVES Increasing obesity rates and accelerating climate change represent two global health challenges shaped by lifestyle change and human environmental modifications. Yet, few studies have considered how these issues may interact to exacerbate disease risk. METHODS In this theory article, we explore evidence that obesity-related disease and climatic changes share socio-ecological drivers and may interact to increase human morbidity and mortality risks. Additionally, we consider how obesity-climate change interactions may disproportionately affect vulnerable populations and how anthropological research can be applied to address this concern. RESULTS Interactions between heat stress and cardiometabolic disease represent an important pathway through which climate change and obesity-related morbidities may jointly impair health. For example, individuals with higher body fatness and obesity-related metabolic conditions (eg, type 2 diabetes) exhibit a reduced ability to dissipate heat. The risk of poor health resulting from these interactions is expected to be heterogeneous, with low- and middle-income countries, individuals of lower socioeconomic status, and minority populations facing a greater disease burden due to relative lack of resource access (eg, air conditioning). Moreover, older adults are at higher risk due to aging-associated changes in body composition and loss of thermoregulation capabilities. CONCLUSIONS Few policy makers appear to be considering how interventions can be designed to simultaneously address the medical burden posed by increasing obesity rates and climate change. Anthropological research is well situated to address this need in a nuanced and culturally-sensitive way; producing research that can be used to support community resilience, promote holistic well-being, and improve health outcomes.
Collapse
Affiliation(s)
- Theresa E Gildner
- Department of Anthropology, Dartmouth College, Hanover, New Hampshire, USA
| | - Stephanie B Levy
- Department of Anthropology, Hunter College, New York, New York, USA.,New York Consortium in Evolutionary Primatology, New York, New York, USA
| |
Collapse
|
31
|
Hussain MF, Roesler A, Kazak L. Regulation of adipocyte thermogenesis: mechanisms controlling obesity. FEBS J 2020; 287:3370-3385. [PMID: 32301220 DOI: 10.1111/febs.15331] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/26/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Adipocyte biology has been intensely researched in recent years due to the emergence of obesity as a serious global health concern and because of the realization that adipose tissue is more than simply a cell type that stores and releases lipids. The plasticity of adipose tissues, to rapidly adapt to altered physiological states of energy demand, is under neuronal and endocrine control. The capacity for white adipocytes to store chemical energy in lipid droplets is key for protecting other organs from the toxic effects of ectopic lipid deposition. In contrast, thermogenic (brown and beige) adipocytes combust macronutrients to generate heat. The thermogenic activity of adipocytes allows them to protect themselves and other tissues from lipid overaccumulation. Advances in brown fat biology have uncovered key molecular players involved in adipocyte determination, differentiation, and thermogenic activation. It is now, well appreciated that three distinct adipocyte types exist: white, beige, and brown. Moreover, functional differences are present within adipocyte subtypes located in anatomically distinct locations. Adding to this complexity is the recent realization from single-cell sequencing studies that adipocyte progenitors are also heterogeneous. Understanding the molecular details of how to increase the number of thermogenic fat cells and their activation may delineate some of the pathophysiological basis of obesity and obesity-related diseases. Here, we review recent advances that have extended our understanding of the central role that adipose tissue plays in energy balance and the mechanisms that control their amount and function.
Collapse
Affiliation(s)
- Mohammed Faiz Hussain
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Anna Roesler
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Lawrence Kazak
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| |
Collapse
|
32
|
Bast-Habersbrunner A, Fromme T. Purine Nucleotides in the Regulation of Brown Adipose Tissue Activity. Front Endocrinol (Lausanne) 2020; 11:118. [PMID: 32210919 PMCID: PMC7076073 DOI: 10.3389/fendo.2020.00118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/24/2020] [Indexed: 01/06/2023] Open
Abstract
Non-shivering thermogenesis in mammalian brown adipose tissue is a powerful mechanism to defend normothermia in cold climates. To minimize the loss of chemical energy, the central functional component, mitochondrial uncoupling protein 1, UCP1, must be tightly regulated. The canonical pathway of UCP1 activation includes lipolytic release of free fatty acids in response to an adrenergic signal. Activating fatty acids overcome constitutive inhibition of UCP1 by the di- and triphosphate forms of purine nucleotides, i.e., ATP, ADP, GTP, and GDP. Cellular concentrations of inhibitory, free nucleotides are subject to significant, adrenergically induced alterations. The regulatory components involved may constitute novel drug targets to manipulate brown fat thermogenesis and thereby organismic energy balance. We here review evidence for and against a dominant role of nucleotides in thermogenic control, describe conceptual routes to endogenously and pharmacologically alter free nucleotide pool size, speculate on a signaling role of degradation products released from active brown fat, and highlight gaps in our understanding of signaling and metabolic pathways involved.
Collapse
Affiliation(s)
- Andrea Bast-Habersbrunner
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Munich, Germany
| | - Tobias Fromme
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Munich, Germany
- *Correspondence: Tobias Fromme
| |
Collapse
|