1
|
Shatarat AT, Shurrab AM, Al-Lahham HM, Tarboush NA, Badran DH, Salameh MA, Badran R. Irisin Modulates Perivascular Adipose Tissue Structure In Rat Thoracic Aorta. Morphologie 2025; 109:100955. [PMID: 39985838 DOI: 10.1016/j.morpho.2025.100955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Irisin is a newly identified hormone secreted mainly by skeletal muscles. It has different effects on the cardiovascular system and blood vessels. The present study investigated the possible effects of irisin on the microscopic structure of the perivascular adipose tissue in the rat thoracic aorta. MATERIALS AND METHODS Twenty rats were randomly divided into two groups: a non-injected control group (n=4) and four injected experimental groups (each n=4). The experimental rats were injected intraperitoneally with one of the following concentrations of irisin; 250, 500, 1000, and 2000ng/mL, twice a week for 4 weeks. After that, all experimental rats' descending thoracic aortas were removed, and imaging was performed. ANOVA and Bonferroni's Multiple Comparison Test were used to achieve statistical comparisons. RESULTS A trend of a dose-dependent increase in the number of brown adipocytes in all irisin-injected groups reached statistical significance at a dose of 2000ng/mL, compared to that of the control group (from 7.9±1 control to 67±6.1 in 2000ng/mL of irisin). A dose-dependent decrease in the number of white adipocytes compared to that of the control group (from 40±4.8 control to 3±0.9 in 2000ng/mL of irisin). CONCLUSION The present study has for the first time demonstrated that irisin has significantly increased the number of brown adipocytes and decreased the number of white adipocytes in the perivascular adipose tissue in rat thoracic aorta.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/anatomy & histology
- Aorta, Thoracic/cytology
- Fibronectins/pharmacology
- Fibronectins/administration & dosage
- Rats
- Male
- Adipose Tissue/drug effects
- Dose-Response Relationship, Drug
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/cytology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/cytology
- Rats, Wistar
- Random Allocation
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- A T Shatarat
- Aqaba Medical Sciences University, Aqaba, Jordan
| | - A M Shurrab
- Al-Balqa Applied University, Al-Salt, Jordan.
| | | | | | - D H Badran
- Ibn Sina University for Medical Sciences, Amman, Jordan
| | - M A Salameh
- Al-Balqa Applied University, Al-Salt, Jordan
| | - R Badran
- Philadelphia University, Amman, Jordan
| |
Collapse
|
2
|
Blasco-Roset A, Quesada-López T, Mestres-Arenas A, Villarroya J, Godoy-Nieto FJ, Cereijo R, Rupérez C, Neess D, Færgeman NJ, Giralt M, Planavila A, Villarroya F. Acyl CoA-binding protein in brown adipose tissue acts as a negative regulator of adaptive thermogenesis. Mol Metab 2025; 96:102153. [PMID: 40220929 PMCID: PMC12050000 DOI: 10.1016/j.molmet.2025.102153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025] Open
Abstract
OBJECTIVE Defective activity of brown adipose tissue (BAT) is linked to obesity and cardiometabolic diseases. While much is known regarding the biological signals that trigger BAT thermogenesis, relatively little is known about the repressors that may impair BAT function in physiological and pathological settings. Acyl CoA-binding protein (ACBP; also known as diazepam binding inhibitor, DBI) has intracellular functions related to lipid metabolism and can be secreted to act as a circulating regulatory factor that affects multiple organs. Our objective was to determine the role of ACBP in BAT function. METHODS Experimental models based on the targeted inactivation of the Acbp gene in brown adipocytes, both in vitro and in vivo, as well as brown adipocytes treated with recombinant ACBP, were developed and analyzed for transcriptomic and metabolic changes. RESULTS ACBP expression and release in BAT are suppressed by noradrenergic cAMP-dependent signals that stimulate thermogenesis. This regulation occurs through gene expression modulation and autophagy-related processes. Mice with targeted ablation of Acbp in brown adipocytes exhibit enhanced BAT thermogenic activity and protection against high-fat diet-induced obesity and glucose intolerance; this is associated with BAT transcriptome changes, including upregulation of BAT thermogenesis-related genes. Treatment of brown adipocytes with exogenous ACBP suppresses oxidative activity, lipolysis, and thermogenesis-related gene expression. ACBP treatment inhibits the noradrenergic-induced phosphorylation of p38 MAP-kinase and CREB, which are major intracellular mediators of brown adipocyte thermogenesis. CONCLUSIONS The ACBP system acts as a crucial auto regulatory repressor of BAT thermogenesis that responds reciprocally to the noradrenergic induction of BAT activity.
Collapse
Affiliation(s)
- Albert Blasco-Roset
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Institut de Recerca de Sant Joan de Déu, 08028 Barcelona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, 28029 Madrid, Spain
| | - Tania Quesada-López
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Institut de Recerca de Sant Joan de Déu, 08028 Barcelona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, 28029 Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB-SANT PAU), Barcelona, Spain; Department of Infectious Diseases, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - Alberto Mestres-Arenas
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Institut de Recerca de Sant Joan de Déu, 08028 Barcelona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, 28029 Madrid, Spain
| | - Joan Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Institut de Recerca de Sant Joan de Déu, 08028 Barcelona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, 28029 Madrid, Spain
| | - Francisco J Godoy-Nieto
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Institut de Recerca de Sant Joan de Déu, 08028 Barcelona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, 28029 Madrid, Spain
| | - Rubén Cereijo
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Institut de Recerca de Sant Joan de Déu, 08028 Barcelona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, 28029 Madrid, Spain
| | - Celia Rupérez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Institut de Recerca de Sant Joan de Déu, 08028 Barcelona, Spain
| | - Ditte Neess
- Department of Biochemistry and Molecular Biology, University of South Denmark, DK-5230, Odense, Denmark
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, University of South Denmark, DK-5230, Odense, Denmark
| | - Marta Giralt
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Institut de Recerca de Sant Joan de Déu, 08028 Barcelona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, 28029 Madrid, Spain
| | - Anna Planavila
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Institut de Recerca de Sant Joan de Déu, 08028 Barcelona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, 28029 Madrid, Spain
| | - Francesc Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Institut de Recerca de Sant Joan de Déu, 08028 Barcelona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, 28029 Madrid, Spain.
| |
Collapse
|
3
|
Wang Y, Fu J, He W, Gao Y, Du J, Xu J, Guo L, Liu Y. Bone marrow mesenchymal stem cells ameliorate diet-induced obesity by activating thermogenesis and alleviating inflammation in adipose tissue. Biochem Biophys Res Commun 2025; 747:151172. [PMID: 39793396 DOI: 10.1016/j.bbrc.2024.151172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025]
Abstract
Obesity and its related metabolic disorders seriously threaten our health and significantly reduce our life expectancy. The aim of the present study was to explore the effects of bone marrow mesenchymal stem cells (BMSCs) on high-fat diet (HFD)-induced obesity mice. The results demonstrated that BMSCs significantly reduced body weight, improved glucose tolerance and insulin sensitivity in obese mice. Further analysis showed that BMSCs could promote brown adipose tissue (BAT) activity and white adipose tissue (WAT) browning by increasing the expression of mitochondrial uncouple protein 1 (UCP1). Additionally, BMSCs markedly increase mitochondrial biogenesis, activate oxidative phosphorylation (OXPHOS) in adipose tissue, further contributing to energy metabolism regulation. Moreover, BMSCs were effective in inhibiting macrophage-related inflammation in adipose tissue, thereby mitigating obesity-associated inflammatory responses. Overall, our results lay the foundation for research on the potential of BMSCs as a promising strategy in alleviating obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Yanxue Wang
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, PR China.
| | - Jingfei Fu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, PR China.
| | - Wanghong He
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, PR China.
| | - Yike Gao
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, PR China.
| | - Juan Du
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, PR China.
| | - Junji Xu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, PR China.
| | - Lijia Guo
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, PR China; Department of Orthodontics (WangFuJing Campus), School of Stomatology, Capital Medical University, Beijing, PR China.
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, PR China.
| |
Collapse
|
4
|
Rezq S, Huffman AM, Basnet J, Alsemeh AE, do Carmo JM, Yanes Cardozo LL, Romero DG. MicroRNA-21 modulates brown adipose tissue adipogenesis and thermogenesis in a mouse model of polycystic ovary syndrome. Biol Sex Differ 2024; 15:53. [PMID: 38987854 PMCID: PMC11238487 DOI: 10.1186/s13293-024-00630-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 06/26/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS), the most common endocrine disorder in premenopausal women, is associated with increased obesity, hyperandrogenism, and altered brown adipose tissue (BAT) thermogenesis. MicroRNAs play critical functions in brown adipocyte differentiation and maintenance. We aim to study the role of microRNA-21 (miR-21) in altered energy homeostasis and BAT thermogenesis in a PCOS mouse model of peripubertal androgen exposure. METHODS Three-week-old miR-21 knockout (miR21KO) or wild-type (WT) female mice were treated with dihydrotestosterone (DHT) or vehicle for 90 days. Body composition was determined by EchoMRI. Energy expenditure (EE), oxygen consumption (VO2), carbon dioxide production (VCO2), and respiratory exchange ratio (RER) were measured by indirect calorimetry. Androgen receptor (AR), and markers of adipogenesis, de novo lipogenesis, angiogenesis, extracellular matrix remodeling, and thermogenesis were quantified by RT-qPCR and/or Western-blot. RESULTS MiR-21 ablation attenuated DHT-mediated increase in body weight while having no effect on fat or BAT mass. MiR-21 ablation attenuated DHT-mediated BAT AR upregulation. MiR-21 ablation did not alter EE; however, miR21KO DHT-treated mice have reduced VO2, VCO2, and RER. MiR-21 ablation reversed DHT-mediated decrease in food intake and increase in sleep time. MiR-21 ablation decreased some adipogenesis (Adipoq, Pparγ, and Cebpβ) and extracellular matrix remodeling (Mmp-9 and Timp-1) markers expression in DHT-treated mice. MiR-21 ablation abolished DHT-mediated increases in thermogenesis markers Cpt1a and Cpt1b, while decreasing CIDE-A expression. CONCLUSIONS Our findings suggest that BAT miR-21 may play a role in regulating DHT-mediated thermogenic dysfunction in PCOS. Modulation of BAT miR-21 levels could be a novel therapeutic approach for the treatment of PCOS-associated metabolic derangements.
Collapse
Affiliation(s)
- Samar Rezq
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
| | - Alexandra M Huffman
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Jelina Basnet
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Amira E Alsemeh
- Department of Anatomy, Histology, and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Jussara M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Licy L Yanes Cardozo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Medicine, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Damian G Romero
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
5
|
Wang Q, Su Y, Sun R, Xiong X, Guo K, Wei M, Yang G, Ru Y, Zhang Z, Li J, Zhang J, Qiao Q, Li X. MIIP downregulation drives colorectal cancer progression through inducing peri-cancerous adipose tissue browning. Cell Biosci 2024; 14:12. [PMID: 38245780 PMCID: PMC10800076 DOI: 10.1186/s13578-023-01179-0] [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] [Accepted: 12/05/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND The enrichment of peri-cancerous adipose tissue is a distinctive feature of colorectal cancer (CRC), accelerating disease progression and worsening prognosis. The communication between tumor cells and adjacent adipocytes plays a crucial role in CRC advancement. However, the precise regulatory mechanisms are largely unknown. This study aims to explore the mechanism of migration and invasion inhibitory protein (MIIP) downregulation in the remodeling of tumor cell-adipocyte communication and its role in promoting CRC. RESULTS MIIP expression was found to be decreased in CRC tissues and closely associated with adjacent adipocyte browning. In an in vitro co-culture model, adipocytes treated with MIIP-downregulated tumor supernatant exhibited aggravated browning and lipolysis. This finding was further confirmed in subcutaneously allografted mice co-injected with adipocytes and MIIP-downregulated murine CRC cells. Mechanistically, MIIP interacted with the critical lipid mobilization factor AZGP1 and regulated AZGP1's glycosylation status by interfering with its association with STT3A. MIIP downregulation promoted N-glycosylation and over-secretion of AZGP1 in tumor cells. Subsequently, AZGP1 induced adipocyte browning and lipolysis through the cAMP-PKA pathway, releasing free fatty acids (FFAs) into the microenvironment. These FFAs served as the primary energy source, promoting CRC cell proliferation, invasion, and apoptosis resistance, accompanied by metabolic reprogramming. In a tumor-bearing mouse model, inhibition of β-adrenergic receptor or FFA uptake, combined with oxaliplatin, significantly improved therapeutic efficacy in CRC with abnormal MIIP expression. CONCLUSIONS Our data demonstrate that MIIP plays a regulatory role in the communication between CRC and neighboring adipose tissue by regulating AZGP1 N-glycosylation and secretion. MIIP reduction leads to AZGP1 oversecretion, resulting in adipose browning-induced CRC rapid progression and poor prognosis. Inhibition of β-adrenergic receptor or FFA uptake, combined with oxaliplatin, may represent a promising therapeutic strategy for CRC with aberrant MIIP expression.
Collapse
Affiliation(s)
- Qinhao Wang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Yuanyuan Su
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
- Department of Pharmacology, Medical College, Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Ruiqi Sun
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Xin Xiong
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Kai Guo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Mengying Wei
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Guodong Yang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yi Ru
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhengxiang Zhang
- Department of Pharmacology, Medical College, Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Jing Li
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jing Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Medicine, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Qing Qiao
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| | - Xia Li
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
6
|
Shen M, Zhang M, Mao N, Lin Z. Batokine in Central Nervous System Diseases. Mol Neurobiol 2023; 60:7021-7031. [PMID: 37526894 DOI: 10.1007/s12035-023-03490-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/06/2023] [Indexed: 08/02/2023]
Abstract
Brown adipose tissue (BAT) is a special type of fat tissue in mammals and is also a key endocrine organ in the human body. Batokine, the endocrine effector of BAT, plays a neuroprotective role and improves the prognosis by exerting anti-apoptotic and anti-inflammatory effects, as well as by improving vascular endothelial function and other mechanisms in nerve injury diseases. The present article briefly reviewed several types of batokines related to central nervous system (CNS) diseases. Following this, the potential therapeutic value and future research direction of batokines for CNS diseases were chiefly discussed from the aspects of protective mechanism and signaling pathway.
Collapse
Affiliation(s)
- Ming Shen
- Department of Neonatology, The Second Affiliated Hospital of Wenzhou Medical University and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang, China
| | - Min Zhang
- Department of Neonatology, The Second Affiliated Hospital of Wenzhou Medical University and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang, China
| | - Niping Mao
- Department of Neonatology, The Second Affiliated Hospital of Wenzhou Medical University and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang, China
| | - Zhenlang Lin
- Department of Neonatology, The Second Affiliated Hospital of Wenzhou Medical University and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China.
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China.
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang, China.
| |
Collapse
|
7
|
Torequl Islam M, Shimul Bhuia M, Paulo Martins de Lima J, Paulo Araujo Maia F, Beatriz Herminia Ducati A, Douglas Melo Coutinho H. Phytanic acid, an inconclusive phytol metabolite: A review. Curr Res Toxicol 2023; 5:100120. [PMID: 37744206 PMCID: PMC10515296 DOI: 10.1016/j.crtox.2023.100120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/28/2023] [Accepted: 08/30/2023] [Indexed: 09/26/2023] Open
Abstract
Phytanic acid (PA: 3,7,11,15-tetramethylhexadecanoic acid) is an important biometabolite of the chlorophyll-derived diterpenoid phytol. Its biological sources (occurrence) and ADME (absorption, distribution, metabolism, and elimination) profile are well-discussed in the literature. Cumulative literature suggests that PA has beneficial as well as harmful biological roles in humans and other animals. This study aimed to sketch a brief summary of PA's beneficial and harmful pharmacological effects in test systems on the basis of existing literature reports. Literature findings propose that PA has anti-inflammatory and immunomodulatory, antidiabetic, anti-obesity, anticancer, and oocyte maturation effects. Although a high plasma PA-level mediated SLS remains controversial, it is evident to link it with Refsum's disease and other peroxisomal enzyme deficiency diseases in humans, including RCDP and LD; ZHDA and Alzheimer's disease; progressive ataxia and dysarthria; and an increased risk of some lymphomas such as LBL, FL, and NHL. PA exerts toxic effects on different kinds of cells, including neuronal, cardiac, and renal cells, through diverse pathways such as oxidative stress, mitochondrial disturbance, apoptosis, disruption of Na+/K+-ATPase activity, Ca2+ homeostasis, alteration of AChE and MAO activities, etc. PA is considered a cardiac biomarker in humans. In conclusion, PA may be one of the most important biometabolites in humans.
Collapse
Affiliation(s)
- Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Md. Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | | | | | | | | |
Collapse
|
8
|
Gangitano E, Martinez-Sanchez N, Bellini MI, Urciuoli I, Monterisi S, Mariani S, Ray D, Gnessi L. Weight Loss and Sleep, Current Evidence in Animal Models and Humans. Nutrients 2023; 15:3431. [PMID: 37571368 PMCID: PMC10420950 DOI: 10.3390/nu15153431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Sleep is a vital process essential for survival. The trend of reduction in the time dedicated to sleep has increased in industrialized countries, together with the dramatic increase in the prevalence of obesity and diabetes. Short sleep may increase the risk of obesity, diabetes and cardiovascular disease, and on the other hand, obesity is associated with sleep disorders, such as obstructive apnea disease, insomnia and excessive daytime sleepiness. Sleep and metabolic disorders are linked; therefore, identifying the physiological and molecular pathways involved in sleep regulation and metabolic homeostasis can play a major role in ameliorating the metabolic health of the individual. Approaches aimed at reducing body weight could provide benefits for both cardiometabolic risk and sleep quality, which indirectly, in turn, may determine an amelioration of the cardiometabolic phenotype of individuals. We revised the literature on weight loss and sleep, focusing on the mechanisms and the molecules that may subtend this relationship in humans as in animal models.
Collapse
Affiliation(s)
- Elena Gangitano
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Noelia Martinez-Sanchez
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
| | | | - Irene Urciuoli
- Department of Surgery, Sapienza University of Rome, 00161 Rome, Italy
| | - Stefania Monterisi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Stefania Mariani
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - David Ray
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
| | - Lucio Gnessi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
9
|
Laiglesia LM, Escoté X, Sáinz N, Felix-Soriano E, Santamaría E, Collantes M, Fernández-Galilea M, Colón-Mesa I, Martínez-Fernández L, Quesada-López T, Quesada-Vázquez S, Rodríguez-Ortigosa C, Arbones-Mainar JM, Valverde ÁM, Martínez JA, Dalli J, Herrero L, Lorente-Cebrián S, Villarroya F, Moreno-Aliaga MJ. Maresin 1 activates brown adipose tissue and promotes browning of white adipose tissue in mice. Mol Metab 2023; 74:101749. [PMID: 37271337 PMCID: PMC10331312 DOI: 10.1016/j.molmet.2023.101749] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 05/19/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023] Open
Abstract
OBJECTIVE Maresin 1 (MaR1) is a docosahexaenoic acid-derived proresolving lipid mediator with insulin-sensitizing and anti-steatosis properties. Here, we aim to unravel MaR1 actions on brown adipose tissue (BAT) activation and white adipose tissue (WAT) browning. METHODS MaR1 actions were tested in cultured murine brown adipocytes and in human mesenchymal stem cells (hMSC)-derived adipocytes. In vivo effects of MaR1 were tested in diet-induced obese (DIO) mice and lean WT and Il6 knockout (Il6-/-) mice. RESULTS In cultured differentiated murine brown adipocytes, MaR1 reduces the expression of inflammatory genes, while stimulates glucose uptake, fatty acid utilization and oxygen consumption rate, along with the upregulation of mitochondrial mass and genes involved in mitochondrial biogenesis and function and the thermogenic program. In Leucine Rich Repeat Containing G Protein-Coupled Receptor 6 (LGR6)-depleted brown adipocytes using siRNA, the stimulatory effect of MaR1 on thermogenic genes was abrogated. In DIO mice, MaR1 promotes BAT remodeling, characterized by higher expression of genes encoding for master regulators of mitochondrial biogenesis and function and iBAT thermogenic activation, together with increased M2 macrophage markers. In addition, MaR1-treated DIO mice exhibit a better response to cold-induced BAT activation. Moreover, MaR1 induces a beige adipocyte signature in inguinal WAT of DIO mice and in hMSC-derived adipocytes. MaR1 potentiates Il6 expression in brown adipocytes and BAT of cold exposed lean WT mice. Interestingly, the thermogenic properties of MaR1 were abrogated in Il6-/- mice. CONCLUSIONS These data reveal MaR1 as a novel agent that promotes BAT activation and WAT browning by regulating thermogenic program in adipocytes and M2 polarization of macrophages. Moreover, our data suggest that LGR6 receptor is mediating MaR1 actions on brown adipocytes, and that IL-6 is required for the thermogenic effects of MaR1.
Collapse
Affiliation(s)
- Laura M Laiglesia
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain
| | - Xavier Escoté
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain; Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, 43204 Spain
| | - Neira Sáinz
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain
| | - Elisa Felix-Soriano
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain
| | - Eva Santamaría
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid 28029, Spain; Division of Hepatology, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - María Collantes
- Department of Nuclear Medicine/ Translational Molecular Imaging Unit (UNIMTRA), Clínica Universidad de Navarra, Pamplona, 31008, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Marta Fernández-Galilea
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Ignacio Colón-Mesa
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain
| | - Leyre Martínez-Fernández
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain
| | - Tania Quesada-López
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine of the University of Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | | | - José M Arbones-Mainar
- Adipocyte and Fat Biology Laboratory (AdipoFat), Instituto de Investigación Sanitaria Aragón, Instituto Aragonés de Ciencias de la Salud, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Ángela M Valverde
- Alberto Sols Biomedical Research Institute (IIBm) (CSIC/UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - J Alfredo Martínez
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK; Center for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| | - Laura Herrero
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Silvia Lorente-Cebrián
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain; Current address: Department of Pharmacology, Physiology, Legal and Forensic Medicine. Faculty of Health and Sport Science, University of Zaragoza, Zaragoza, Spain
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine of the University of Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María J Moreno-Aliaga
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| |
Collapse
|
10
|
Zhao L, Zhou J, Chen J, Zhang X, Zhang H, Guo L, Li D, Ning J, Wang X, Jin W, Mai K, Abraham E, Butcher R, Sun J. A chemical signal that promotes insect survival via thermogenesis. RESEARCH SQUARE 2023:rs.3.rs-2756320. [PMID: 37214941 PMCID: PMC10197781 DOI: 10.21203/rs.3.rs-2756320/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Cold-activated thermogenesis of brown adipose tissues (BAT) is vital for the survival of animals under cold stress and also inhibits the development of tumours. The development of small-molecule tools that target thermogenesis pathways could lead to novel therapies against cold, obesity, and even cancer. Here, we identify a chemical signal that is produced in beetles in the winter to activate fat thermogenesis. This hormone elevates the basal body temperature by increasing cellular mitochondrial density and uncoupling in order to promote beetle survival. We demonstrate that this hormone activates UCP4- mediated uncoupled respiration through adipokinetic hormone receptor (AKHR). This signal serves as a novel fat-burning activator that utilizes a conserved mechanism to promote thermogenesis not only in beetles, nematode and flies, but also in mice, protecting the mice against cold and tumor growth. This hormone represents a new strategy to manipulate fat thermogenesis.
Collapse
Affiliation(s)
- Lilin Zhao
- Institute of Zoology, Chinese Academy of Sciences
| | - Jiao Zhou
- Institute of Zoology, Chinese Academy of Sciences
| | - Junxian Chen
- Institute of Zoology, Chinese Academy of Sciences
| | | | | | | | - Defeng Li
- Institute of Microbiology, Chinese Academy of Sciences
| | - Jing Ning
- Institute of Zoology, Chinese Academy of Sciences
| | - Xinchen Wang
- Institute of Zoology, Chinese Academy of Sciences
| | - Wanzhu Jin
- Institute of Zoology, Chinese Academy of Sciences
| | - Kevin Mai
- Department of Chemistry, University of Florida
| | | | | | | |
Collapse
|
11
|
Castellá M, Villarroya F. Brown fat. Med Clin (Barc) 2023:S0025-7753(23)00050-7. [PMID: 36868963 DOI: 10.1016/j.medcli.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 03/05/2023]
Affiliation(s)
- Moisés Castellá
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, España; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, España
| | - Francesc Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, España; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, España.
| |
Collapse
|
12
|
Latteri S, Sofia M, Puleo S, Di Vincenzo A, Cinti S, Castorina S. Mechanisms linking bariatric surgery to adipose tissue, glucose metabolism, fatty liver disease and gut microbiota. Langenbecks Arch Surg 2023; 408:101. [PMID: 36826628 PMCID: PMC9957865 DOI: 10.1007/s00423-023-02821-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 01/25/2023] [Indexed: 02/25/2023]
Abstract
PURPOSE In the last 20 years, bariatric surgery has achieved an important role in translational and clinical research because of obesity comorbidities. Initially, a tool to lose weight, bariatric surgery now has been shown to be involved in several metabolic pathways. METHODS We conducted a narrative review discussing the underlying mechanisms that could explain the impact of bariatric surgery and the relationship between obesity and adipose tissue, T2D, gut microbiota, and NAFLD. RESULTS Bariatric surgery has an impact in the relation between obesity and type 2 diabetes, but in addition it induces the white-to-brown adipocyte trans-differentiation, by enhancing thermogenesis. Another issue is the connection of bariatric surgery with the gut microbiota and its role in the complex mechanism underlying weight gain. CONCLUSION Bariatric surgery modifies gut microbiota, and these modifications influence lipid metabolism, leading to improvement of non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Saverio Latteri
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Maria Sofia
- Department of General Surgery, Cannizzaro Hospital, Via Messina 829, 95126, Catania, Italy.
| | - Stefano Puleo
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| | - Angelica Di Vincenzo
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto 10A, 60020, Ancona, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto 10A, 60020, Ancona, Italy
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| |
Collapse
|
13
|
Pan J, Kothan S, Moe Moe AT, Huang K. Dysfunction of insulin-AKT-UCP1 signalling inhibits transdifferentiation of human and mouse white preadipocytes into brown-like adipocytes. Adipocyte 2022; 11:213-226. [PMID: 35416120 PMCID: PMC9009895 DOI: 10.1080/21623945.2022.2062852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 02/07/2023] Open
Abstract
The mechanism of insulin signaling on browning of white preadipocytes remains unclear. Human and mouse primary subcutaneous white preadipocytes (hsASCs and WT lean and obese msASCs, respectively) were induced to transdifferentiate into beige adipocytes under conditions of intact or blocked insulin signaling, respectively. Level of phosphoinositide-3-kinase (PI3K) after induction of beige adipocytes under conditions of normal insulin signaling, phosphorylated protein kinase B (pAKT), peroxisome proliferator-activated receptor γ coactivator-1 alpha (PGC-1α), zinc-fifinger transcriptional factor PRD1-BF1-RIZ1 homologous domain-containing protein 16 (PRDM16), uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT/enhancer binding protein beta (C/EBPβ) were significantly increased. Conversely, when insulin signaling is incompletely inhibited, the expression of the thermogenic and adipogenic factors is significantly reduced, with obvious impairment of adipogenesis. However, phosphorylation level of adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) and expression level of sirtuin type 1 (SIRT1) had increased. These white preadipocytes from different donors showed similar dynamic change in morphology and molecular levels during the browning. The present data indicate that insulin signaling plays a important role in regulation of browning of hsASCs and msASCs through PI3K-AKT-UCP1 signaling pathway. The insulin-AMPK-SIRT1 pathway was also involved in the adipocytes browning, while its effect is limited.
Collapse
Affiliation(s)
- Jie Pan
- Shandong Provincial Key Laboratory of Animal Resistant Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong Province, China
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Suchart Kothan
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Aye Thidar Moe Moe
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Kun Huang
- Shandong Provincial Key Laboratory of Animal Resistant Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong Province, China
| |
Collapse
|
14
|
Rial-Pensado E, Rivas-Limeres V, Grijota-Martínez C, Rodríguez-Díaz A, Capelli V, Barca-Mayo O, Nogueiras R, Mittag J, Diéguez C, López M. Temperature modulates systemic and central actions of thyroid hormones on BAT thermogenesis. Front Physiol 2022; 13:1017381. [PMID: 36467699 PMCID: PMC9716276 DOI: 10.3389/fphys.2022.1017381] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/03/2022] [Indexed: 08/07/2023] Open
Abstract
Thyroid hormones (THs) play a major role regulating energy balance and brown adipose tissue (BAT) thermogenesis, as well as body temperature, as shown in hyperthyroid patients. However, the current landscape of preclinical thyroid hormone models is complex. For example, while rats become catabolic after TH administration, mice gain weight; so, these differences in species need to be analyzed in detail and specially whether temperature could be a factor. Here, we aimed to investigate the effect of environmental temperature on those actions. Rats were subcutaneously treated with L-thyroxine (T4) or stereotaxically within the ventromedial nucleus of the hypothalamus (VMH) with triiodothyronine (T3) and housed at 23°C, 4°C or 30°C; energy balance, BAT thermogenesis and AMP-activated protein kinase (AMPK) in the VMH were analyzed. Our data showed that the effect of both systemic T4 of central T3 on energy balance and BAT thermogenesis was dependent upon environmental temperature. This evidence is of interest in the design of experimental settings highlighting the species-specific metabolic actions of THs, and in understanding its physiological role in the adaptation to temperature.
Collapse
Affiliation(s)
- Eva Rial-Pensado
- Department of Physiology, CIMUS, University of Santiago de Compostela- Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Verónica Rivas-Limeres
- Department of Physiology, CIMUS, University of Santiago de Compostela- Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Carmen Grijota-Martínez
- Department of Cell Biology, Faculty of Biology, Complutense University, Madrid, Spain
- Alberto Sols Biomedical Research Institute (CSIC-UAM), Madrid, Spain
| | - Amanda Rodríguez-Díaz
- Department of Physiology, CIMUS, University of Santiago de Compostela- Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Valentina Capelli
- Department of Physiology, CIMUS, University of Santiago de Compostela- Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Olga Barca-Mayo
- Department of Physiology, CIMUS, University of Santiago de Compostela- Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela- Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Jens Mittag
- Institute for Endocrinology and Diabetes—Molecular Endocrinology, Center of Brain Behavior and Metabolism CBBM, University of Lübeck, Lübeck, Germany
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela- Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela- Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| |
Collapse
|
15
|
Bertola A, Gallerand A, Ivanov S. Immune cell involvement in brown adipose tissue functions. DISCOVERY IMMUNOLOGY 2022; 1:kyac007. [PMID: 38566905 PMCID: PMC10917225 DOI: 10.1093/discim/kyac007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/15/2022] [Accepted: 10/28/2022] [Indexed: 04/04/2024]
Abstract
Brown adipose tissue (BAT) contains many immune cells. The presence of macrophages, monocytes, dendritic cells, T cells, B cells, and mast cells was documented in BAT. However, in comparison to white adipose tissue, relatively little is known on the impact of immune cells on BAT function. By directly interacting with BAT stromal cells, or by secreting pro- and anti-inflammatory mediators, immune cells modulate BAT activation and subsequently influence on adaptative thermogenesis and heat generation. In the current manuscript, we will focus on the diversity and functions of BAT immune cells.
Collapse
|
16
|
Alizadeh H. Meteorin-like protein (Metrnl): A metabolic syndrome biomarker and an exercise mediator. Cytokine 2022; 157:155952. [PMID: 35780711 DOI: 10.1016/j.cyto.2022.155952] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/04/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022]
Abstract
Metrnl is a secreted protein able to activate different intracellular signaling pathways in adipocytes, macrophages, myocytes and cardiomyocytes with physiological effects of the browning of white adipose tissue (BWT), insulin sensitivity, inflammation inhibition, skeletal muscle regeneration and heart protection. Shown to be regulated by obesity, diabetes, caloric restriction, weight loss and heart diseases, Metrnl is definitely involved in metabolic turbulences, and may play roles in metabolic syndrome (MetS). However, due to the conflicting data yielded, Metrnl is still far from clinical application as a diagnostic and/or a therapeutic agent or even a therapeutic target in MetS-related diseases such as type 2 diabetes (T2D) and obesity. Nevertheless, blood Metrnl levels as well as Metrnl as a cardiokine have been reported to play cardioprotective roles against heart diseases. Considering the established metabolic and anti-inflammatory hallmarks, exercise-induced Metrnl (as a myokine) is regarded as an exercise mediator in improving obesity-induced complications such as insulin resistance, T2D and inflammation. Besides, due to its healing role in muscle damage, Metrnl is also a potential therapeutic candidate to enhance regeneration with ageing or other inflammatory myopathies like Duchenne muscular dystrophy (DMD). Therefore, there are still many exercise-related questions unanswered on Metrnl, such as Metrnl-mediated fat browning in humans, exercise effects on heart Metrnl production and secretion and the effects of other exercise-induced skeletal muscle stressors like hypoxia and oxidative in Metrnl production other than exercise-induced muscle damage.
Collapse
Affiliation(s)
- Hamid Alizadeh
- Exercise Physiology, University of Mazandaran, Babolsar, Mazandaran, Iran.
| |
Collapse
|
17
|
Yoshida Y, Shimizu I, Hsiao YT, Suda M, Katsuumi G, Seki M, Suzuki Y, Okuda S, Soga T, Minamino T. Differing impact of phosphoglycerate mutase 1-deficiency on brown and white adipose tissue. iScience 2022; 25:104268. [PMID: 35521515 PMCID: PMC9065309 DOI: 10.1016/j.isci.2022.104268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/03/2022] [Accepted: 04/13/2022] [Indexed: 11/27/2022] Open
Abstract
Brown adipose tissue (BAT) is a metabolically active organ that contributes to the thermogenic response to cold exposure. In addition, other thermogenic cells termed beige adipocytes are generated in white adipose tissue (WAT) by cold exposure. Although activation of brown/beige adipose tissue is associated with mobilization of both glucose and lipids, few studies have focused on the role of glycolytic enzymes in regulating adipose tissue function. We generated mouse models with specific deletion of the glycolytic enzyme phosphoglycerate mutase 1 (PGAM1) from adipose tissue. Deletion of Pgam1 from both BAT and WAT promoted whitening of BAT with beiging of visceral WAT, whereas deletion of Pgam1 from BAT alone led to whitening of BAT without beiging of WAT. Our results demonstrate a potential role of glycolytic enzymes in beiging of visceral WAT and suggest that PGAM1 would be a novel therapeutic target in obesity and diabetes. Pgam1 deletion leads to whitening of brown adipose tissue Pgam1 deletion promotes beiging of visceral white adipose tissue (WAT) Pgam1 deletion-induced beiging is associated with increased levels of amino acids
Collapse
Affiliation(s)
- Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.,Department of Advanced Senotherapeutics, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yung-Ting Hsiao
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata 997-0052, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| |
Collapse
|
18
|
Kihara-Negishi F, Ohkura N, Takahashi Y, Fujita T, Nakamura Y, Maruyama K, Oshitari T, Yamaguchi S. Nobiletin and 3′-Demethyl Nobiletin Activate Brown Adipocytes upon β-Adrenergic Stimulation. Biol Pharm Bull 2022; 45:528-533. [DOI: 10.1248/bpb.b21-00988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
19
|
Johansen OS, Ma T, Gerhart-Hines Z. Leveraging GPCR signaling in thermogenic fat to counteract metabolic diseases. Mol Metab 2022; 60:101474. [PMID: 35339729 PMCID: PMC9046952 DOI: 10.1016/j.molmet.2022.101474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/24/2022] [Accepted: 03/11/2022] [Indexed: 11/05/2022] Open
Abstract
Background Thermogenic brown and beige adipocytes are recognized for their unique capacity to consume extraordinary levels of metabolites and lipids from the blood to fuel heat-producing catabolic processes [[1], [2], [3], [4], [5], [6], [7]]. In humans, the functions of thermogenic adipocytes are associated with cardiometabolic protection and improved glycemic control [[8], [9], [10], [11], [12], [13]]. Consequently, engaging these macronutrient-consuming and energy-dissipating activities has gained attention as a promising therapeutic strategy for counteracting metabolic diseases, such as obesity and diabetes. Scope of review In this review, we highlight new advances in our understanding of the physiological role of G protein-coupled receptors (GPCRs) in controlling thermogenic adipocyte biology. We further extend our discussion to the opportunities and challenges posed by pharmacologically targeting different elements of GPCR signaling in these highly specialized fat cells. Major conclusions GPCRs represent appealing candidates through which to harness adipose thermogenesis. Yet safely and effectively targeting these druggable receptors on brown and beige adipocytes has thus far proven challenging. Therefore, continued interrogation across the GPCR landscape is necessary for future leaps within the field of thermogenic fat biology to unlock the therapeutic potential of adipocyte catabolism. Brown and beige thermogenic adipocytes robustly consume and catabolize macronutrients. The catabolic activity of thermogenic adipocytes promotes organismal energy balance. Thermogenic adipocyte functions are tightly controlled by G protein-coupled receptors (GPCRs). GPCRs can be potentially targeted at multiple levels to therapeutically harness thermogenic activity.
Collapse
Affiliation(s)
- Olivia Sveidahl Johansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, DK
| | - Tao Ma
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, DK; Embark Biotech ApS, Copenhagen, DK
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, DK; Embark Biotech ApS, Copenhagen, DK; Center for Adipocyte Signaling, Odense, DK.
| |
Collapse
|
20
|
PET/MRI-evaluated brown adipose tissue activity may be related to dietary MUFA and omega-6 fatty acids intake. Sci Rep 2022; 12:4112. [PMID: 35260768 PMCID: PMC8904502 DOI: 10.1038/s41598-022-08125-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/03/2022] [Indexed: 12/17/2022] Open
Abstract
An investigation of new ways to activate brown adipose tissue (BAT) is highly valuable, as it is a possible tool for obesity prevention and treatment. The aim of our study was to evaluate the relationships between dietary intake and BAT activity. The study group comprised 28 healthy non-smoking males aged 21–42 years. All volunteers underwent a physical examination and 75-g OGTT and completed 3-day food intake diaries to evaluate macronutrients and fatty acid intake. Body composition measurements were assessed using DXA scanning. An FDG-18 PET/MR was performed to visualize BAT activity. Brown adipose tissue was detected in 18 subjects (67% normal-weight individuals and 33% overweight/obese). The presence of BAT corresponded with a lower visceral adipose tissue (VAT) content (p = 0.04, after adjustment for age, daily kcal intake, and DXA Lean mass). We noted significantly lower omega-6 fatty acids (p = 0.03) and MUFA (p = 0.02) intake in subjects with detected BAT activity after adjustment for age, daily average kcal intake, and DXA Lean mass, whereas omega-3 fatty acids intake was comparable between the two groups. BAT presence was positively associated with the concentration of serum IL-6 (p = 0.01) during cold exposure. Our results show that BAT activity may be related to daily omega-6 fatty acids intake.
Collapse
|
21
|
Gupta A, Shamsi F, Altemose N, Dorlhiac GF, Cypess AM, White AP, Yosef N, Patti ME, Tseng YH, Streets A. Characterization of transcript enrichment and detection bias in single-nucleus RNA-seq for mapping of distinct human adipocyte lineages. Genome Res 2022; 32:242-257. [PMID: 35042723 PMCID: PMC8805720 DOI: 10.1101/gr.275509.121] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 12/10/2021] [Indexed: 02/02/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) enables molecular characterization of complex biological tissues at high resolution. The requirement of single-cell extraction, however, makes it challenging for profiling tissues such as adipose tissue, for which collection of intact single adipocytes is complicated by their fragile nature. For such tissues, single-nucleus extraction is often much more efficient and therefore single-nucleus RNA sequencing (snRNA-seq) presents an alternative to scRNA-seq. However, nuclear transcripts represent only a fraction of the transcriptome in a single cell, with snRNA-seq marked with inherent transcript enrichment and detection biases. Therefore, snRNA-seq may be inadequate for mapping important transcriptional signatures in adipose tissue. In this study, we compare the transcriptomic landscape of single nuclei isolated from preadipocytes and mature adipocytes across human white and brown adipocyte lineages, with whole-cell transcriptome. We show that snRNA-seq is capable of identifying the broad cell types present in scRNA-seq at all states of adipogenesis. However, we also explore how and why the nuclear transcriptome is biased and limited, as well as how it can be advantageous. We robustly characterize the enrichment of nuclear-localized transcripts and adipogenic regulatory lncRNAs in snRNA-seq, while also providing a detailed understanding for the preferential detection of long genes upon using this technique. To remove such technical detection biases, we propose a normalization strategy for a more accurate comparison of nuclear and cellular data. Finally, we show successful integration of scRNA-seq and snRNA-seq data sets with existing bioinformatic tools. Overall, our results illustrate the applicability of snRNA-seq for the characterization of cellular diversity in the adipose tissue.
Collapse
Affiliation(s)
- Anushka Gupta
- University of California at Berkeley-University of California at San Francisco Graduate Program in Bioengineering, Berkeley, California 94720, USA
| | - Farnaz Shamsi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Nicolas Altemose
- University of California at Berkeley-University of California at San Francisco Graduate Program in Bioengineering, Berkeley, California 94720, USA
| | - Gabriel F Dorlhiac
- Biophysics Graduate Group, University of California at Berkeley, Berkeley, California 94720, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Andrew P White
- Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Nir Yosef
- Center for Computational Biology, University of California, Berkeley, Berkeley, California 94720, USA
- Department of Electrical Engineering and Computer Sciences, University of California at Berkeley, Berkeley, California 94720, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts 02139, USA
| | | | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Aaron Streets
- University of California at Berkeley-University of California at San Francisco Graduate Program in Bioengineering, Berkeley, California 94720, USA
- Biophysics Graduate Group, University of California at Berkeley, Berkeley, California 94720, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| |
Collapse
|
22
|
Bonfante ILP, Duft RG, Mateus KCDS, Trombeta JCDS, Finardi EAR, Ramkrapes APB, Brunelli DT, Mori MADS, Chacon-Mikahil MPT, Velloso LA, Cavaglieri CR. Acute/Chronic Responses of Combined Training on Serum Pro-thermogenic/Anti-inflammatory Inducers and Its Relation With Fed and Fasting State in Overweight Type 2 Diabetic Individuals. Front Physiol 2022; 12:736244. [PMID: 35126168 PMCID: PMC8811167 DOI: 10.3389/fphys.2021.736244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Concentrations of pro-thermogenic/anti-inflammatory inductors are influenced by fed/fasting, sedentary/trained states, and metabolic pattern. However, there is a lack of information on the interactions of these conditions, especially in humans. Thus, the present study aimed to evaluate the chronic and acute training responses as well as the fed/fasted states of serum pro-thermogenic/anti-inflammatory inducers in overweight type 2 diabetics individuals. Fifteen individuals with type 2 diabetes [body mass index (BMI): 29.61 ± 3.60 kg/m2; age: 50.67 ± 3.97 years] participated in the study. In the pre- and post-experimental periods, baseline clinical parameters analyses were performed. Pro-thermogenic/anti-inflammatory inductors were evaluated pre/post-baseline and before, shortly after, and after 30′ and 60′ in the first and last sessions of a 16-week combined training (CT) period. These inducers were also compared for fasting and feeding before and after the training period. CT has improved baseline physical fitness, metabolic pattern, and it has also increased interleukin (IL)33 and FNDC5/irisin. In the first training session, there was a decrease in IL4, IL13, and IL33, besides an increase in FNDC5/irisin, and natriuretic peptides. In the last training session, there was an increase in natriuretic peptides and bone morphogenic protein 4 (BMP4). Differences in responses between the first and last training sessions were observed at certain post-session times for IL4, IL33, and natriuretic peptides, always with higher concentrations occurring in the last session. In evaluating the area under the curve (AUC) of the first and last training session, FNDC5/irisin, natriuretics peptides, and meteorin-like showed increased areas in the last training session. The pre-training fed state showed an increase in IL4 and IL33, while in fasting there was an increase in meteorin-like, natriuretic peptides, and FNDC5/irisin. In the post-training, IL4, IL13, and IL33 were increased in the fed state, while meteorin-like, natriuretic peptides, and FNDC5/irisin remained increased in the fast. Adaptation to physical training and a better metabolic pattern favor an improvement in the acute secretory pattern in part of pro-thermogenic and anti-inflammatory substances analyzed. The fed and fasting states also interfere differently in these substances, where fasting interferes with the increase of myokines, while the fed state induces an increase in interleukins.Clinical Trial Registration: [http://www.ensaiosclinicos.gov.br/rg/RBR-62n5qn/], identifier [U1111-1202-1476].
Collapse
Affiliation(s)
- Ivan Luiz Padilha Bonfante
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, Campinas, Brazil
- Federal Institute of Education, Science and Technology of São Paulo, Hortolândia Campus, Hortolândia, Brazil
- *Correspondence: Ivan Luiz Padilha Bonfante,
| | - Renata Garbellini Duft
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, Campinas, Brazil
| | | | | | | | - Ana Paula Boito Ramkrapes
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, Campinas, Brazil
| | - Diego Trevisan Brunelli
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, Campinas, Brazil
| | - Marcelo Alves da Silva Mori
- Laboratory of Aging Biology, Department of Biochemistry and Tissue Biology, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | | | - Licio Augusto Velloso
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, Brazil
| | - Cláudia Regina Cavaglieri
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, Campinas, Brazil
| |
Collapse
|
23
|
Luo J, Wang Y, Gilbert E, Liu D. Deletion of GPR30 Drives the Activation of Mitochondrial Uncoupling Respiration to Induce Adipose Thermogenesis in Female Mice. Front Endocrinol (Lausanne) 2022; 13:877152. [PMID: 35592783 PMCID: PMC9110859 DOI: 10.3389/fendo.2022.877152] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Thermogenic adipocytes possess a promising approach to combat obesity with its capability promoting energy metabolism. We previously discovered that deletion of GPR30 (GPRKO), a presumably membrane-associated estrogen receptor, protected female mice from developing obesity, glucose intolerance, and insulin resistance when challenged with a high-fat diet (HFD). In vivo, the metabolic phenotype of wild type (WT) and GPRKO female mice were measured weekly. Acute cold tolerance test was performed. Ex vivo, mitochondrial respiration of brown adipose tissue (BAT) was analyzed from diet-induced obese female mice of both genotypes. In vitro, stromal vascular fractions (SVF) were isolated for beige adipocyte differentiation to investigate the role of GPR30 in thermogenic adipocyte. Deletion of GPR30 protects female mice from hypothermia and the mitochondria in BAT are highly energetic in GPRKO animals while the WT mitochondria remain in a relatively quiescent stage. Consistently, GPR30 deficiency enhances beige adipocyte differentiation in white adipose tissue (WAT) and activates the thermogenic browning of subcutaneous WAT due to up-regulation of UCP-1, which thereby protects female mice from HFD-induced obesity. GPR30 is a negative regulator of thermogenesis, which at least partially contributes to the reduced adiposity in the GPRKO female mice. Our findings provide insight into the mechanism by which GPR30 regulates fat metabolism and adiposity in female mice exposed to excess calories, which may be instrumental in the development of new therapeutic strategies for obesity.
Collapse
Affiliation(s)
- Jing Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
- Department of Human Nutrition, Foods and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Yao Wang
- Department of Human Nutrition, Foods and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Elizabeth Gilbert
- Department of Animal and Poultry Sciences, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Dongmin Liu
- Department of Human Nutrition, Foods and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA, United States
- *Correspondence: Dongmin Liu,
| |
Collapse
|
24
|
miR-21 mimic blocks obesity in mice: A novel therapeutic option. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 26:401-416. [PMID: 34552821 PMCID: PMC8426473 DOI: 10.1016/j.omtn.2021.06.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/25/2021] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) are promising drug targets for obesity and metabolic disorders. Recently, miRNA mimics are providing a unique mechanism of action that guides the process for drug development and sets out the context of their therapeutic application. miRNA (miR)-21 expression in white adipose tissue (WAT) has been associated with obesity. We aimed to analyze miR-21 expression levels in relation to diabetes and obesity to determine the effect that miR-21 mimic has on processes involved in WAT functionality, to dissect the underlying molecular mechanisms, and to study the potential therapeutic application of the miR-21 mimic against obesity. We found higher miR-21 levels in WAT from non-diabetic obese compared to normoweight humans and mice. Moreover, in 3T3-L1 adipocytes, miR-21 mimic affect genes involved in WAT functionality regulation and significantly increase the expression of genes involved in browning and thermogenesis. Interestingly, in vivo treatment with the miR-21 mimic blocked weight gain induced by a high-fat diet in obese mice, without modifying food intake or physical activity. This was associated with metabolic enhancement, WAT browning, and brown adipose tissue (AT) thermogenic programming through vascular endothelial growth factor A (VEGF-A), p53, and transforming growth factor β1 (TGF-β1) signaling pathways. Our findings suggest that miR-21 mimic-based therapy may provide a new opportunity to therapeutically manage obesity and consequently, its associated alterations.
Collapse
|
25
|
Theobromine enhances the conversion of white adipocytes into beige adipocytes in a PPARγ activation-dependent manner. J Nutr Biochem 2021; 100:108898. [PMID: 34748921 DOI: 10.1016/j.jnutbio.2021.108898] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/07/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022]
Abstract
The adipocytes play an important role in driving the obese-state-white adipose tissue (WAT) stores the excess energy as fat, wherein brown adipose tissue (BAT) is responsible for energy expenditure via the thermoregulatory function of uncoupling protein 1 (UCP1)-the imbalance between these two onsets obesity. Moreover, the anti-obesity effects of brown-like-adipocytes (beige) in WAT are well documented. Browning, the process of transformation of energy-storing into energy-dissipating adipocytes, is a potential preventive strategy against obesity and its related diseases. In the present study, to explore an alternative source of natural products in the regulation of adipocyte transformation, we assessed the potential of theobromine (TB), a bitter alkaloid of the cacao plant, inducing browning in mice (in vivo) and primary adipocytes (in vitro). Dietary supplementation of TB significantly increased skin temperature of the inguinal region in mice and induced the expression of UCP1 protein. It also increased the expression levels of mitochondrial marker proteins in subcutaneous adipose tissues but not in visceral adipose tissues. The microarray analysis showed that TB supplementation upregulated multiple thermogenic and beige adipocyte marker genes in subcutaneous adipose tissue. Furthermore, in mouse-derived primary adipocytes, TB upregulated the expression of the UCP1 protein and mitochondrial mass in a PPARγ ligand-dependent manner. It also increased the phosphorylation levels of PPARγ coactivator 1α without affecting its protein expression. These results indicate that dietary supplementation of TB induces browning in subcutaneous WAT and enhances PPARγ-induced UCP1 expression in vitro, suggesting its potential to treat obesity.
Collapse
|
26
|
FGF2 disruption enhances thermogenesis in brown and beige fat to protect against adiposity and hepatic steatosis. Mol Metab 2021; 54:101358. [PMID: 34710640 PMCID: PMC8605413 DOI: 10.1016/j.molmet.2021.101358] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
Objective Fibroblast growth factor 2 (FGF2) has been reported to play divergent roles in white adipogenic differentiation, however, whether it regulates thermogenesis of fat tissues remains largely unknown. We therefore aimed to investigate the effect of FGF2 on fat thermogenesis and elucidate the underlying mechanisms. Methods FGF2-KO and wild-type (WT) mice were fed with chow diet and high-fat diet (HFD) for 14 weeks. The brown and white fat mass, thermogenic capability, respiratory exchange ratio, and hepatic fat deposition were determined. In vitro experiments were conducted to compare the thermogenic ability of FGF2-KO- with WT-derived brown and white adipocytes. Exogenous FGF2 was supplemented to in vitro-cultured WT brown and ISO-induced beige adipocytes. The FGFR inhibitor, PPARγ agonist, and PGC-1α expression lentivirus were used with the aid of technologies including Co-IP, ChIP, and luciferase reporter assay to elucidate the mechanisms underlying the FGF2 regulation of thermogenesis. Results FGF2 gene disruption results in increased thermogenic capability in both brown and beige fat, supporting by increased UCP1 expression, enhanced respiratory exchange ratio, and elevated thermogenic potential in response to cold exposure. Thus, the deletion of FGF2 protects mice from high fat-induced adiposity and hepatic steatosis. Mechanistically, in vitro investigations indicated FGF2 acts in autocrine/paracrine fashions. Exogenous FGF2 supplementation inhibits both PGC-1α and PPARγ expression, leading to suppression of UCP1 expression in brown and beige adipocytes. Conclusions These findings demonstrate that FGF2 is a novel thermogenic regulator, suggesting a viable potential strategy for using FGF2-selective inhibitors in combat adiposity and associated hepatic steatosis. FGF2-KO mice show potentiated stimulation on thermogenic capability under both basal and cold challenge stimulation. FGF2 disruption protected mice against HFD-induced adiposity and hepatic steatosis. FGF2 acts in autocrine/paracrine fashions in vitro. Both PPARγ and PGC-1α play roles in FGF2 suppression of thermogenesis.
Collapse
|
27
|
Antoniak K, Hansdorfer-Korzon R, Mrugacz M, Zorena K. Adipose Tissue and Biological Factors. Possible Link between Lymphatic System Dysfunction and Obesity. Metabolites 2021; 11:metabo11090617. [PMID: 34564433 PMCID: PMC8464765 DOI: 10.3390/metabo11090617] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022] Open
Abstract
The World Health Organization (WHO) has recognised obesity as one of the top ten threats to human health. Obesity is not only a state of abnormally increased adipose tissue in the body, but also of an increased release of biologically active metabolites. Moreover, obesity predisposes the development of metabolic syndrome and increases the incidence of type 2 diabetes (T2DM), increases the risk of developing insulin resistance, atherosclerosis, ischemic heart disease, polycystic ovary syndrome, hypertension and cancer. The lymphatic system is a one-directional network of thin-walled capillaries and larger vessels covered by a continuous layer of endothelial cells that provides a unidirectional conduit to return filtered arterial and tissue metabolites towards the venous circulation. Recent studies have shown that obesity can markedly impair lymphatic function. Conversely, dysfunction in the lymphatic system may also be involved in the pathogenesis of obesity. This review highlights the important findings regarding obesity related to lymphatic system dysfunction, including clinical implications and experimental studies. Moreover, we present the role of biological factors in the pathophysiology of the lymphatic system and we propose the possibility of a therapy supporting the function of the lymphatic system in the course of obesity.
Collapse
Affiliation(s)
- Klaudia Antoniak
- Department of Immunobiology and Environment Microbiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland;
| | - Rita Hansdorfer-Korzon
- Department of Physical Therapy, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland;
| | - Małgorzata Mrugacz
- Department of Ophthalmology and Eye Rehabilitation, Medical University of Bialystok, Kilinskiego 1, 15-089 Białystok, Poland;
| | - Katarzyna Zorena
- Department of Immunobiology and Environment Microbiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland;
- Correspondence: ; Tel./Fax: +48-583491765
| |
Collapse
|
28
|
Abstract
The classical dogma states that brown adipose tissue (BAT) plays a major role in the regulation of temperature in neonates. However, although BAT has been studied in infants for more than a century, the knowledge about its physiological features at this stage of life is rather limited. This has been mainly due to the lack of appropriate investigation methods, ethically suitable for neonates. Here, we have applied non-invasive infrared thermography (IRT) to investigate neonatal BAT activity. Our data show that BAT temperature correlates with body temperature and that mild cold stimulus promotes BAT activation in newborns. Notably, a single short-term cold stimulus during the first day of life improves the body temperature adaption to a subsequent cold event. Finally, we identify that bone morphogenic protein 8B (BMP8B) is associated with the BAT thermogenic response in neonates. Overall, our data uncover key features of the setup of BAT thermogenesis in newborns.
Collapse
|
29
|
Sheng Y, Xia F, Chen L, Lv Y, Lv S, Yu J, Liu J, Ding G. Differential Responses of White Adipose Tissue and Brown Adipose Tissue to Calorie Restriction During Aging. J Gerontol A Biol Sci Med Sci 2021; 76:393-399. [PMID: 32222773 DOI: 10.1093/gerona/glaa070] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Indexed: 01/15/2023] Open
Abstract
Age-related adipose tissue dysfunction is potentially important in the development of insulin resistance and metabolic disorder. Caloric restriction (CR) is a robust intervention to reduce adiposity, improve metabolic health, and extend healthy life span. Both white adipose tissue (WAT) and brown adipose tissue (BAT) are involved in energy homeostasis. CR triggers the beiging of WAT in young mice; however, the effects of CR on beiging of WAT and function of BAT during aging are unclear. This study aimed to investigate how age and CR impact the beiging of WAT, the function of BAT, and metabolic health in mice. C57BL/6 mice were fed CR diet (40% less than the ad libitum [AL] diet) for 3 months initiated in young (3 months), middle-aged (12 months), and old (19 months) stage. We found age-related changes in different types of adipose tissue, including adipocyte enlargement, declined beiging of WAT, and declined thermogenic and β-oxidational function of BAT. Moreover, CR attenuated age-associated adipocyte enlargement and prevented the age-related decline in beiging potential of WAT. These protective effects on the beiging potential were significant in inguinal WAT at all three ages, which were significant in epididymal WAT at young and old age. In contrast, thermogenic and β-oxidational function of BAT further declined after CR in the young age group. In conclusion, our findings reveal the contribution of WAT beiging decline to age-related metabolic disorder and suggest nutritional intervention, specifically targeting WAT beiging, as an effective approach to metabolic health during aging.
Collapse
Affiliation(s)
- Yunlu Sheng
- Division of Geriatric Endocrinology, the First Affiliated Hospital of Nanjing Medical University, People's Republic of China
| | - Fan Xia
- Division of Geriatric Endocrinology, the First Affiliated Hospital of Nanjing Medical University, People's Republic of China
| | - Lei Chen
- Division of Geriatric Respiratory, the First Affiliated Hospital of Nanjing Medical University, People's Republic of China
| | - Yifan Lv
- Division of Geriatric Endocrinology, the First Affiliated Hospital of Nanjing Medical University, People's Republic of China
| | - Shan Lv
- Division of Geriatric Endocrinology, the First Affiliated Hospital of Nanjing Medical University, People's Republic of China
| | - Jing Yu
- Division of Geriatric Endocrinology, the First Affiliated Hospital of Nanjing Medical University, People's Republic of China
| | - Juan Liu
- Division of Geriatric Endocrinology, the First Affiliated Hospital of Nanjing Medical University, People's Republic of China
| | - Guoxian Ding
- Division of Geriatric Endocrinology, the First Affiliated Hospital of Nanjing Medical University, People's Republic of China
| |
Collapse
|
30
|
Calvo E, Keiran N, Núñez-Roa C, Maymó-Masip E, Ejarque M, Sabadell-Basallote J, Del Mar Rodríguez-Peña M, Ceperuelo-Mallafré V, Seco J, Benaiges E, Michalopoulou T, Jorba R, Vendrell J, Fernández-Veledo S. Effects of stem cells from inducible brown adipose tissue on diet-induced obesity in mice. Sci Rep 2021; 11:13923. [PMID: 34230537 PMCID: PMC8260805 DOI: 10.1038/s41598-021-93224-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/16/2021] [Indexed: 11/11/2022] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) are a promising option for the treatment of obesity and its metabolic co-morbidities. Despite the recent identification of brown adipose tissue (BAT) as a potential target in the management of obesity, the use of ASCs isolated from BAT as a therapy for patients with obesity has not yet been explored. Metabolic activation of BAT has been shown to have not only thermogenic effects, but it also triggers the secretion of factors that confer protection against obesity. Herein, we isolated and characterized ASCs from the visceral adipose tissue surrounding a pheochromocytoma (IB-hASCs), a model of inducible BAT in humans. We then compared the anti-obesity properties of IB-hASCs and human ASCs isolated from visceral white adipose tissue (W-hASCs) in a murine model of diet-induced obesity. We found that both ASC therapies mitigated the metabolic abnormalities of obesity to a similar extent, including reducing weight gain and improving glucose tolerance. However, infusion of IB-hASCs was superior to W-hASCs in suppressing lipogenic and inflammatory markers, as well as preserving insulin secretion. Our findings provide evidence for the metabolic benefits of visceral ASC infusion and support further studies on IB-hASCs as a therapeutic option for obesity-related comorbidities.
Collapse
Affiliation(s)
- Enrique Calvo
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain
| | - Noelia Keiran
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira I Virgili, Tarragona, Spain
| | - Catalina Núñez-Roa
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain
| | - Elsa Maymó-Masip
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain
| | - Miriam Ejarque
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Sabadell-Basallote
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira I Virgili, Tarragona, Spain
| | - María Del Mar Rodríguez-Peña
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain
| | - Victòria Ceperuelo-Mallafré
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira I Virgili, Tarragona, Spain
| | - Jesús Seco
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain
| | - Ester Benaiges
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira I Virgili, Tarragona, Spain
| | - Theodora Michalopoulou
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain
| | - Rosa Jorba
- Servei de Cirurgia General I de L'Aparell Digestiu, Hospital Universitari Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Joan Vendrell
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain.
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain.
- Universitat Rovira I Virgili, Tarragona, Spain.
| | - Sonia Fernández-Veledo
- Servei D'Endocrinologia I Nutrició I Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut D'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch 4, 43007, Tarragona, Spain.
- CIBER de Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM) - Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
31
|
Sveidahl Johansen O, Ma T, Hansen JB, Markussen LK, Schreiber R, Reverte-Salisa L, Dong H, Christensen DP, Sun W, Gnad T, Karavaeva I, Nielsen TS, Kooijman S, Cero C, Dmytriyeva O, Shen Y, Razzoli M, O'Brien SL, Kuipers EN, Nielsen CH, Orchard W, Willemsen N, Jespersen NZ, Lundh M, Sustarsic EG, Hallgren CM, Frost M, McGonigle S, Isidor MS, Broholm C, Pedersen O, Hansen JB, Grarup N, Hansen T, Kjær A, Granneman JG, Babu MM, Calebiro D, Nielsen S, Rydén M, Soccio R, Rensen PCN, Treebak JT, Schwartz TW, Emanuelli B, Bartolomucci A, Pfeifer A, Zechner R, Scheele C, Mandrup S, Gerhart-Hines Z. Lipolysis drives expression of the constitutively active receptor GPR3 to induce adipose thermogenesis. Cell 2021; 184:3502-3518.e33. [PMID: 34048700 PMCID: PMC8238500 DOI: 10.1016/j.cell.2021.04.037] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 02/10/2021] [Accepted: 04/23/2021] [Indexed: 12/19/2022]
Abstract
Thermogenic adipocytes possess a therapeutically appealing, energy-expending capacity, which is canonically cold-induced by ligand-dependent activation of β-adrenergic G protein-coupled receptors (GPCRs). Here, we uncover an alternate paradigm of GPCR-mediated adipose thermogenesis through the constitutively active receptor, GPR3. We show that the N terminus of GPR3 confers intrinsic signaling activity, resulting in continuous Gs-coupling and cAMP production without an exogenous ligand. Thus, transcriptional induction of Gpr3 represents the regulatory parallel to ligand-binding of conventional GPCRs. Consequently, increasing Gpr3 expression in thermogenic adipocytes is alone sufficient to drive energy expenditure and counteract metabolic disease in mice. Gpr3 transcription is cold-stimulated by a lipolytic signal, and dietary fat potentiates GPR3-dependent thermogenesis to amplify the response to caloric excess. Moreover, we find GPR3 to be an essential, adrenergic-independent regulator of human brown adipocytes. Taken together, our findings reveal a noncanonical mechanism of GPCR control and thermogenic activation through the lipolysis-induced expression of constitutively active GPR3.
Collapse
Affiliation(s)
- Olivia Sveidahl Johansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Embark Biotech ApS, Copenhagen, Denmark; Center for Adipocyte Signaling, University of Southern Denmark, Odense, Denmark
| | - Tao Ma
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Embark Biotech ApS, Copenhagen, Denmark
| | - Jakob Bondo Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Embark Biotech ApS, Copenhagen, Denmark
| | - Lasse Kruse Markussen
- Center for Adipocyte Signaling, University of Southern Denmark, Odense, Denmark; Functional Genomics and Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Laia Reverte-Salisa
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Hua Dong
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | | | - Wenfei Sun
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Thorsten Gnad
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Iuliia Karavaeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Svava Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Cheryl Cero
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Oksana Dmytriyeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Yachen Shen
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Shannon L O'Brien
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK; Institute of Pharmacology and Toxicology and Bio-Imaging Center, University of Würzburg, Würzburg, Germany
| | - Eline N Kuipers
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Carsten Haagen Nielsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen, Denmark
| | | | - Nienke Willemsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Naja Zenius Jespersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Morten Lundh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Elahu Gosney Sustarsic
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie Mørch Hallgren
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Frost
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Seth McGonigle
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Marie Sophie Isidor
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Christa Broholm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Bo Hansen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Kjær
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen, Denmark
| | - James G Granneman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - M Madan Babu
- MRC Laboratory of Molecular Biology, Cambridge, UK; Department of Structural Biology and Center for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Davide Calebiro
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK; Institute of Pharmacology and Toxicology and Bio-Imaging Center, University of Würzburg, Würzburg, Germany
| | - Søren Nielsen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Raymond Soccio
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jonas Thue Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Thue Walter Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Embark Biotech ApS, Copenhagen, Denmark
| | - Brice Emanuelli
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Mandrup
- Center for Adipocyte Signaling, University of Southern Denmark, Odense, Denmark; Functional Genomics and Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Embark Biotech ApS, Copenhagen, Denmark; Center for Adipocyte Signaling, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
32
|
Nascimento EBM, Moonen MPB, Remie CME, Gariani K, Jörgensen JA, Schaart G, Hoeks J, Auwerx J, van Marken Lichtenbelt WD, Schrauwen P. Nicotinamide Riboside Enhances In Vitro Beta-adrenergic Brown Adipose Tissue Activity in Humans. J Clin Endocrinol Metab 2021; 106:1437-1447. [PMID: 33524145 DOI: 10.1210/clinem/dgaa960] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 12/11/2022]
Abstract
CONTEXT Elevating nicotinamide adenine dinucleotide (NAD+) levels systemically improves metabolic health, which can be accomplished via nicotinamide riboside (NR). Previously, it was demonstrated that NR supplementation in high-fat-diet (HFD)-fed mice decreased weight gain, normalized glucose metabolism, and enhanced cold tolerance. OBJECTIVE Because brown adipose tissue (BAT) is a major source of thermogenesis, we hypothesize that NR stimulates BAT in mice and humans. DESIGN AND INTERVENTION HFD-fed C56BL/6J mice were supplemented with 400 mg/kg/day NR for 4 weeks and subsequently exposed to cold. In vitro primary adipocytes derived from human BAT biopsies were pretreated with 50 µM or 500 µM NR before measuring mitochondrial uncoupling. Human volunteers (45-65 years; body mass index, 27-35 kg/m2) were supplemented with 1000 mg/day NR for 6 weeks to determine whether BAT activity increased, as measured by [18F]FDG uptake via positron emission tomography-computed tomography (randomized, double blinded, placebo-controlled, crossover study with NR supplementation). RESULTS NR supplementation in HFD-fed mice decreased adipocyte cell size in BAT. Cold exposure further decreased adipocyte cell size on top of that achieved by NR alone independent of ex vivo lipolysis. In adipocytes derived from human BAT, NR enhanced in vitro norepinephrine-stimulated mitochondrial uncoupling. However, NR supplementation in human volunteers did not alter BAT activity or cold-induced thermogenesis. CONCLUSIONS NR stimulates in vitro human BAT but not in vivo BAT in humans. Our research demonstrates the need for further translational research to better understand the differences in NAD+ metabolism in mouse and human.
Collapse
Affiliation(s)
- Emmani B M Nascimento
- NUTRIM School of Nutrition and Translational Research in Metabolism; Department of Nutrition and Movement Sciences; Maastricht University Medical Center, Maastricht, MD, The Netherlands
| | - Michiel P B Moonen
- NUTRIM School of Nutrition and Translational Research in Metabolism; Department of Nutrition and Movement Sciences; Maastricht University Medical Center, Maastricht, MD, The Netherlands
| | - Carlijn M E Remie
- NUTRIM School of Nutrition and Translational Research in Metabolism; Department of Nutrition and Movement Sciences; Maastricht University Medical Center, Maastricht, MD, The Netherlands
| | - Karim Gariani
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Division of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Geneva University Hospitals, Geneva, Switzerland
| | - Johanna A Jörgensen
- NUTRIM School of Nutrition and Translational Research in Metabolism; Department of Nutrition and Movement Sciences; Maastricht University Medical Center, Maastricht, MD, The Netherlands
| | - Gert Schaart
- NUTRIM School of Nutrition and Translational Research in Metabolism; Department of Nutrition and Movement Sciences; Maastricht University Medical Center, Maastricht, MD, The Netherlands
| | - Joris Hoeks
- NUTRIM School of Nutrition and Translational Research in Metabolism; Department of Nutrition and Movement Sciences; Maastricht University Medical Center, Maastricht, MD, The Netherlands
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Wouter D van Marken Lichtenbelt
- NUTRIM School of Nutrition and Translational Research in Metabolism; Department of Nutrition and Movement Sciences; Maastricht University Medical Center, Maastricht, MD, The Netherlands
| | - Patrick Schrauwen
- NUTRIM School of Nutrition and Translational Research in Metabolism; Department of Nutrition and Movement Sciences; Maastricht University Medical Center, Maastricht, MD, The Netherlands
| |
Collapse
|
33
|
vonderEmbse AN, Elmore SE, Jackson KB, Habecker BA, Manz KE, Pennell KD, Lein PJ, La Merrill MA. Developmental exposure to DDT or DDE alters sympathetic innervation of brown adipose in adult female mice. Environ Health 2021; 20:37. [PMID: 33794904 PMCID: PMC8017793 DOI: 10.1186/s12940-021-00721-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/15/2021] [Indexed: 05/04/2023]
Abstract
BACKGROUND Exposure to the bioaccumulative pesticide dichlorodiphenyltrichloroethane (DDT) and its metabolite dichlorodiphenyldichloroethylene (DDE) has been associated with increased risk of insulin resistance and obesity in humans and experimental animals. These effects appear to be mediated by reduced brown adipose tissue (BAT) thermogenesis, which is regulated by the sympathetic nervous system. Although the neurotoxicity of DDT is well-established, whether DDT alters sympathetic innervation of BAT is unknown. We hypothesized that perinatal exposure to DDT or DDE promotes thermogenic dysfunction by interfering with sympathetic regulation of BAT thermogenesis. METHODS Pregnant C57BL/6 J mice were administered environmentally relevant concentrations of DDTs (p,p'-DDT and o,p'-DDT) or DDE (p,p'-DDE), 1.7 mg/kg and 1.31 mg/kg, respectively, from gestational day 11.5 to postnatal day 5 by oral gavage, and longitudinal body temperature was recorded in male and female offspring. At 4 months of age, metabolic parameters were measured in female offspring via indirect calorimetry with or without the β3 adrenergic receptor agonist, CL 316,243. Immunohistochemical and neurochemical analyses of sympathetic neurons innervating BAT were evaluated. RESULTS We observed persistent thermogenic impairment in adult female, but not male, mice perinatally exposed to DDTs or p,p'-DDE. Perinatal DDTs exposure significantly impaired metabolism in adult female mice, an effect rescued by treatment with CL 316,243 immediately prior to calorimetry experiments. Neither DDTs nor p,p'-DDE significantly altered BAT morphology or the concentrations of norepinephrine and its metabolite DHPG in the BAT of DDTs-exposed mice. However, quantitative immunohistochemistry revealed a 20% decrease in sympathetic axons innervating BAT in adult female mice perinatally exposed to DDTs, but not p,p'-DDE, and 48 and 43% fewer synapses in stellate ganglia of mice exposed to either DDTs or p,p'-DDE, respectively, compared to control. CONCLUSIONS These data demonstrate that perinatal exposure to DDTs or p,p'-DDE impairs thermogenesis by interfering with patterns of connectivity in sympathetic circuits that regulate BAT.
Collapse
Affiliation(s)
- Annalise N. vonderEmbse
- Department of Environmental Toxicology, University of California-Davis College of Agricultural and Environmental Sciences, One Shields Avenue, Davis, CA 95616 USA
- Department of Molecular Biosciences, University of California-Davis, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616 USA
| | - Sarah E. Elmore
- Department of Environmental Toxicology, University of California-Davis College of Agricultural and Environmental Sciences, One Shields Avenue, Davis, CA 95616 USA
- Present address: Office of Environmental Health Hazard Assessment, California EPA, Oakland, CA USA
| | - Kyle B. Jackson
- Department of Environmental Toxicology, University of California-Davis College of Agricultural and Environmental Sciences, One Shields Avenue, Davis, CA 95616 USA
- Integrative Genetics and Genomics Graduate Group, University of California-Davis, Davis, CA USA
| | - Beth A. Habecker
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239 USA
| | - Katherine E. Manz
- School of Engineering, Brown University, 184 Hope Street, Providence, RI 02912 USA
| | - Kurt D. Pennell
- School of Engineering, Brown University, 184 Hope Street, Providence, RI 02912 USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California-Davis, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616 USA
| | - Michele A. La Merrill
- Department of Environmental Toxicology, University of California-Davis College of Agricultural and Environmental Sciences, One Shields Avenue, Davis, CA 95616 USA
| |
Collapse
|
34
|
Yue H, Liu W, Zhang W, Jia M, Huang F, Du F, Xu T. Dietary low ratio of n-6/n-3 polyunsaturated fatty acids improve type 2 diabetes mellitus via activating brown adipose tissue in male mice. J Food Sci 2021; 86:1058-1065. [PMID: 33590526 DOI: 10.1111/1750-3841.15645] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 12/30/2020] [Accepted: 01/19/2021] [Indexed: 12/20/2022]
Abstract
The ratio n-6/n-3 polyunsaturated fatty acids (PUFA) has been caused widespread discussion. However, the best ratio and mechanism of n-6/n-3 PUFA in type 2 diabetes mellitus (T2DM) are largely unknown. This study investigated the effects of different ratio of n-6/n-3 PUFA diets on brown adipose tissue (BAT) and T2DM in mice. Results showed that compared with high ratio of n-6/n-3 PUFA (50:1) diet, lower ratio of n-6/n-3 PUFA (1:1 and 5:1) diets significantly increased BAT mass by 67.55% and 60.49%, decreased the fasting blood glucose (24.87% and 20.64%), total cholesterol (32.9% and 23.84%), triglyceride (33.51% and 29.62%), low-density lipoprotein cholesterol (19.23% and 17.38%), and increased glucose tolerance by 21.99% and 15.52%. Further, qRT-PCR analyses indicated that lower ratio of n-6/n-3 PUFA diets activated BAT, increased the expression of Ucp1, β-3AR, PPAR-γ, cAMP, GLU1, HSL, LPL, and PGC-1α, further improved lipid and glucose metabolism in T2DM mice. In conclusion, this study substantiated that the lower ratio of n-6/n-3 PUFA (1:1 and 5:1) improve symptoms associated with T2DM via activating BAT. PRACTICAL APPLICATION: Dietary ratio of n-6/n-3 polyunsaturated fatty acids is essential for the improvement of chronic diseases. Our current study showed that 1:1 or 5:1 ratio of n-6/n-3 polyunsaturated fatty acids had better efficiency for type 2 diabetes mellitus via activating brown adipose tissue when compared with 1:50. This finding provided useful guidance for the daily diet of patients with diabetes.
Collapse
Affiliation(s)
- Hao Yue
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Shandong Engineering Research Center of Food for Special Medical Purpose/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Wei Liu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Shandong Engineering Research Center of Food for Special Medical Purpose/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Wenlong Zhang
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Shandong Engineering Research Center of Food for Special Medical Purpose/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Min Jia
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Shandong Engineering Research Center of Food for Special Medical Purpose/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Fenghong Huang
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Shandong Engineering Research Center of Food for Special Medical Purpose/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Fangling Du
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Shandong Engineering Research Center of Food for Special Medical Purpose/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Tongcheng Xu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Shandong Engineering Research Center of Food for Special Medical Purpose/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs, Jinan, China
| |
Collapse
|
35
|
Van Schaik L, Kettle C, Green R, Irving HR, Rathner JA. Effects of Caffeine on Brown Adipose Tissue Thermogenesis and Metabolic Homeostasis: A Review. Front Neurosci 2021; 15:621356. [PMID: 33613184 PMCID: PMC7889509 DOI: 10.3389/fnins.2021.621356] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
The impact of brown adipose tissue (BAT) metabolism on understanding energy balance in humans is a relatively new and exciting field of research. The pathogenesis of obesity can be largely explained by an imbalance between caloric intake and energy expenditure, but the underlying mechanisms are far more complex. Traditional non-selective sympathetic activators have been used to artificially elevate energy utilization, or suppress appetite, however undesirable side effects are apparent with the use of these pharmacological interventions. Understanding the role of BAT, in relation to human energy homeostasis has the potential to dramatically offset the energy imbalance associated with obesity. This review discusses paradoxical effects of caffeine on peripheral adenosine receptors and the possible role of adenosine in increasing metabolism is highlighted, with consideration to the potential of central rather than peripheral mechanisms for caffeine mediated BAT thermogenesis and energy expenditure. Research on the complex physiology of adipose tissue, the embryonic lineage and function of the different types of adipocytes is summarized. In addition, the effect of BAT on overall human metabolism and the extent of the associated increase in energy expenditure are discussed. The controversy surrounding the primary β-adrenoceptor involved in human BAT activation is examined, and suggestions as to the lack of translational findings from animal to human physiology and human in vitro to in vivo models are provided. This review compares and distinguishes human and rodent BAT effects, thus developing an understanding of human BAT thermogenesis to aid lifestyle interventions targeting obesity and metabolic syndrome. The focus of this review is on the effect of BAT thermogenesis on overall metabolism, and the potential therapeutic effects of caffeine in increasing metabolism via its effects on BAT.
Collapse
Affiliation(s)
- Lachlan Van Schaik
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
| | - Christine Kettle
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
| | - Rodney Green
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
| | - Helen R. Irving
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
| | - Joseph A. Rathner
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
- Department of Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
36
|
Yao Z, Yan Y, Zheng X, Wang M, Zhang H, Li H, Chen W. Dietary Lactate Supplementation Protects against Obesity by Promoting Adipose Browning in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:14841-14849. [PMID: 33284607 DOI: 10.1021/acs.jafc.0c05899] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Yogurt has been widely used in weight-loss foods to prevent obesity, but its molecular nature remains unclear. Lactate is a major ingredient of yogurt, while its cognate cell surface receptor GPR81 is highly expressed in adipose tissues in mammals. Here we hypothesized that dietary lactate supplementation might activate GPR81 to promote adipose browning. Studying mouse models, we observed that GPR81 was substantially lowered in adipose tissue of obese mice compared with that for lean ones, whereas its expression was markedly up-regulated by a β3-adrenergic receptor (β3-AR) agonist. The deficiency of GPR81 greatly attenuated experimental adipose browning and thermogenesis. Importantly, oral administration of lactate effectively induced adipose browning, enhanced thermogenesis, improved dyslipidemia, and protected mice against high-fat-diet-induced obesity. Mechanistically, p38 mitogen-activated protein kinase might serve as a key downstream effect or of GPR81. Collectively, our findings revealed a critical role of GPR81 in adipose browning and provided a new insight into obesity management by modulating lactate-GPR81 signaling axis.
Collapse
MESH Headings
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Animals
- Dietary Supplements/analysis
- Energy Metabolism/drug effects
- Humans
- Lactic Acid/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Obese
- Obesity/drug therapy
- Obesity/genetics
- Obesity/metabolism
- Obesity/physiopathology
- Protective Agents/administration & dosage
- Receptors, Adrenergic, beta-3/genetics
- Receptors, Adrenergic, beta-3/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
Collapse
Affiliation(s)
- Zhijie Yao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yongheng Yan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xu Zheng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Mingfu Wang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Haitao Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
37
|
Klomklorm A, Ruangthai R, Vaithanomsat P, Sukatta U, Phoemsapthawee J. Concurrent training and Eri silkworm pupae ingestion improve resting and exercise fat oxidation and energy expenditure in obese adults. J Exerc Rehabil 2020; 16:467-479. [PMID: 33178649 PMCID: PMC7609851 DOI: 10.12965/jer.2040682.341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 10/04/2020] [Indexed: 11/29/2022] Open
Abstract
A randomized control trial was conducted to investigate the effects of combined concurrent training and Eri silkworm pupae ingestion on resting and exercise fat oxidation (FAO), as well as energy expenditure, and cardiometabolic risk markers in obese adults. Thirty-six sedentary, obese participants were divided into three groups: (1) placebo control group (CON, n=12), (2) Eri silkworm pupae ingestion group (ERI, n=12), and (3) combined concurrent training and Eri silkworm pupae ingestion group (CBT-ERI, n=12). Participants in the ERI and the CBT-ERI con-sumed 5-g Eri silkworm pupae/day (approximately 2.5-g linolenic acid). The concurrent training program comprised of supervised aerobic and resistance training: three 1-hr sessions/wk for 8 weeks. Body composi-tion, energy expenditure, and FAO at rest and during exercise, heart rate variability, and blood chemistry were measured before and after the 8-week interventions. Following the interventions, resting FAO, the natural logarithm of very low-frequency power (lnVLF), and high-sensi-tive C-reactive protein concentration significantly improved in both the ERI and the CBT-ERI. Only the CBT-ERI improved resting energy expen-diture, FAO during exercise, trunk and gynoid fat mass, total cholesterol concentration, the standard deviation of normal R-R intervals (SDNN), and the percentage difference between adjacent normal R-R intervals >50 ms. Furthermore, there were significant correlations between rest-ing energy expenditure and FAO, lnVLF and SDNN in the CBT-ERI. In conclusion, this study demonstrates that concurrent training together with dietary Eri silkworm pupae leads to increased energy expenditure through a significant increase in FAO at rest and during exercise, as well as reduced fat mass.
Collapse
Affiliation(s)
- Andaman Klomklorm
- Department of Sports Science and Health, Faculty of Sports Science, Kasetsart University, Nakhon Pathom, Thailand
| | - Ratree Ruangthai
- Department of Sports Science and Health, Faculty of Sports Science, Kasetsart University, Nakhon Pathom, Thailand
| | - Pilanee Vaithanomsat
- Kasetsart Agricultural and Agro-Industrial Product Improvement Institute, Kasetsart University, Bangkok, Thailand
| | - Udomlak Sukatta
- Kasetsart Agricultural and Agro-Industrial Product Improvement Institute, Kasetsart University, Bangkok, Thailand
| | - Jatuporn Phoemsapthawee
- Department of Sports Science and Health, Faculty of Sports Science, Kasetsart University, Nakhon Pathom, Thailand
| |
Collapse
|
38
|
Dragano NRV, Fernø J, Diéguez C, López M, Milbank E. Reprint of: Recent Updates on Obesity Treatments: Available Drugs and Future Directions. Neuroscience 2020; 447:191-215. [PMID: 33046217 DOI: 10.1016/j.neuroscience.2020.08.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the last thirty years, obesity has reached epidemic proportions and is now regarded as a major health issue in contemporary society trending to serious economic and social burdens. The latest projections of the World Health Organization are alarming. By 2030, nearly 60% of the worldwide population could be either obese or overweight, highlighting the needs to find innovative treatments. Currently, bariatric surgery is the most effective way to efficiently lower body mass. Although great improvements in terms of recovery and patient care were made in these surgical procedures, bariatric surgery remains an option for extreme forms of obesity and seems unable to tackle obesity pandemic expansion. Throughout the last century, numerous pharmacological strategies targeting either peripheral or central components of the energy balance regulatory system were designed to reduce body mass, some of them reaching sufficient levels of efficiency and safety. Nevertheless, obesity drug therapy remains quite limited on its effectiveness to actually overcome the obesogenic environment. Thus, innovative unimolecular polypharmacology strategies, able to simultaneously target multiple actors involved in the obesity initiation and expansion, were developed during the last ten years opening a new promising avenue in the pharmacological management of obesity. In this review, we first describe the clinical features of obesity-associated conditions and then focus on the outcomes of currently approved drug therapies for obesity as well as new ones expecting to reach the clinic in the near future.
Collapse
Affiliation(s)
- Nathalia R V Dragano
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain.
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Carlos Diéguez
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Miguel López
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Edward Milbank
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain.
| |
Collapse
|
39
|
Seoane-Collazo P, Diéguez C, Nogueiras R, Rahmouni K, Fernández-Real JM, López M. Nicotine' actions on energy balance: Friend or foe? Pharmacol Ther 2020; 219:107693. [PMID: 32987056 DOI: 10.1016/j.pharmthera.2020.107693] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022]
Abstract
Obesity has reached pandemic proportions and is associated with severe comorbidities, such as type 2 diabetes mellitus, hepatic and cardiovascular diseases, and certain cancer types. However, the therapeutic options to treat obesity are limited. Extensive epidemiological studies have shown a strong relationship between smoking and body weight, with non-smokers weighing more than smokers at any age. Increased body weight after smoking cessation is a major factor that interferes with their attempts to quit smoking. Numerous controlled studies in both humans and rodents have reported that nicotine, the main bioactive component of tobacco, exerts a marked anorectic action. Furthermore, nicotine is also known to modulate energy expenditure, by regulating the thermogenic activity of brown adipose tissue (BAT) and the browning of white adipose tissue (WAT), as well as glucose homeostasis. Many of these actions occur at central level, by controlling the activity of hypothalamic neuropeptide systems such as proopiomelanocortin (POMC), or energy sensors such as AMP-activated protein kinase (AMPK). However, direct impact of nicotine on metabolic tissues, such as BAT, WAT, liver and pancreas has also been described. Here, we review the actions of nicotine on energy balance. The relevance of this interaction is interesting, because considering the restricted efficiency of obesity treatments, a possible complementary approach may focus on compounds with known pharmacokinetic profile and pharmacological actions, such as nicotine or nicotinic acetylcholine receptors signaling.
Collapse
Affiliation(s)
- Patricia Seoane-Collazo
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain; International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine and Veterans Affairs Health Care System, Iowa City, IA 52242, USA
| | - José Manuel Fernández-Real
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain; Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain; Department of Diabetes, Endocrinology and Nutrition (UDEN), Hospital of Girona "Dr Josep Trueta" and Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain.
| |
Collapse
|
40
|
Rashid FA, Abbas HJ, Naser NA, Addai Ali H. Effect of Long-Term Moderate Physical Exercise on Irisin between Normal Weight and Obese Men. ScientificWorldJournal 2020; 2020:1897027. [PMID: 32952453 PMCID: PMC7481929 DOI: 10.1155/2020/1897027] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/20/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Irisin is a myokine that has a beneficial effect on obesity and glucose metabolism by increasing energy expenditure. This study aims to investigate the effect of long-term moderate physical exercise on irisin levels and its correlations with body mass index (BMI), waist circumferences (WC), and metabolic parameters in normal weight and obese males. Material and method. A follow-up case-control study of sixty male participants, comprised of thirty normal weight and thirty obese, who had undergone supervised long-term moderate physical exercises for six months. Serum irisin levels, fasting blood glucose, serum insulin, homeostatic model assessment of the insulin resistance index (HOMA-IR), and β-cell function (HOMA-B2) were assessed. RESULTS Long-term moderate exercise induced elevation of the irisin level significantly (P < 0.0001) with significant reduction of the BMI, WC, fasting blood glucose, insulin, HOMA-IR, and HOMA-B2 levels (P < 0.0001) in comparison between obese and normal weight groups. There are significant differences for each parameter in each obese and normal weight group before and after physical exercise with exception of the BMI and WC in the normal group. Significant negative correlations were shown between irisin and blood glucose and insulin and HOMA-IR levels in the obese group and normal weight group. CONCLUSION Irisin improves glucose homeostasis after long-term moderate physical exercises, suggesting that irisin could have regulatory effect on glucose, insulin resistance, and obesity and it could be used as a potential therapy for obesity and insulin resistance.
Collapse
Affiliation(s)
- Farah A. Rashid
- Department of Chemistry, College of Science, Al-Nahrain University, Baghdad, Iraq
| | | | | | - Hana'a Addai Ali
- Department of Chemistry, College of Science, University of Kufa, Najaf, Iraq
| |
Collapse
|
41
|
Nutritional and metabolic regulation of brown and beige adipose tissues. J Physiol Biochem 2020; 76:181-184. [DOI: 10.1007/s13105-020-00745-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/07/2020] [Indexed: 12/24/2022]
|
42
|
Marmol P, Krapacher F, Ibáñez CF. Control of brown adipose tissue adaptation to nutrient stress by the activin receptor ALK7. eLife 2020; 9:54721. [PMID: 32366358 PMCID: PMC7200161 DOI: 10.7554/elife.54721] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/21/2020] [Indexed: 12/22/2022] Open
Abstract
Adaptation to nutrient availability is crucial for survival. Upon nutritional stress, such as during prolonged fasting or cold exposure, organisms need to balance the feeding of tissues and the maintenance of body temperature. The mechanisms that regulate the adaptation of brown adipose tissue (BAT), a key organ for non-shivering thermogenesis, to variations in nutritional state are not known. Here we report that specific deletion of the activin receptor ALK7 in BAT resulted in fasting-induced hypothermia due to exaggerated catabolic activity in brown adipocytes. After overnight fasting, BAT lacking ALK7 showed increased expression of genes responsive to nutrient stress, including the upstream regulator KLF15, aminoacid catabolizing enzymes, notably proline dehydrogenase (POX), and adipose triglyceride lipase (ATGL), as well as markedly reduced lipid droplet size. In agreement with this, ligand stimulation of ALK7 suppressed POX and KLF15 expression in both mouse and human brown adipocytes. Treatment of mutant mice with the glucocorticoid receptor antagonist RU486 restored KLF15 and POX expression levels in mutant BAT, suggesting that loss of BAT ALK7 results in excessive activation of glucocorticoid signaling upon fasting. These results reveal a novel signaling pathway downstream of ALK7 which regulates the adaptation of BAT to nutrient availability by limiting nutrient stress-induced overactivation of catabolic responses in brown adipocytes.
Collapse
Affiliation(s)
- Patricia Marmol
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Favio Krapacher
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Carlos F Ibáñez
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden.,Department of Physiology, National University of Singapore, Singapore, Singapore.,Life Sciences Institute, National University of Singapore, Singapore, Singapore.,Stellenbosch Institute for Advanced Study, Wallenberg Research Centre at Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
43
|
Dragano NRV, Fernø J, Diéguez C, López M, Milbank E. Recent Updates on Obesity Treatments: Available Drugs and Future Directions. Neuroscience 2020; 437:215-239. [PMID: 32360593 DOI: 10.1016/j.neuroscience.2020.04.034] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022]
Abstract
In the last thirty years, obesity has reached epidemic proportions and is now regarded as a major health issue in contemporary society trending to serious economic and social burdens. The latest projections of the World Health Organization are alarming. By 2030, nearly 60% of the worldwide population could be either obese or overweight, highlighting the needs to find innovative treatments. Currently, bariatric surgery is the most effective way to efficiently lower body mass. Although great improvements in terms of recovery and patient care were made in these surgical procedures, bariatric surgery remains an option for extreme forms of obesity and seems unable to tackle obesity pandemic expansion. Throughout the last century, numerous pharmacological strategies targeting either peripheral or central components of the energy balance regulatory system were designed to reduce body mass, some of them reaching sufficient levels of efficiency and safety. Nevertheless, obesity drug therapy remains quite limited on its effectiveness to actually overcome the obesogenic environment. Thus, innovative unimolecular polypharmacology strategies, able to simultaneously target multiple actors involved in the obesity initiation and expansion, were developed during the last ten years opening a new promising avenue in the pharmacological management of obesity. In this review, we first describe the clinical features of obesity-associated conditions and then focus on the outcomes of currently approved drug therapies for obesity as well as new ones expecting to reach the clinic in the near future.
Collapse
Affiliation(s)
- Nathalia R V Dragano
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain.
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Carlos Diéguez
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Miguel López
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Edward Milbank
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain.
| |
Collapse
|
44
|
Yao L, Han Z, Zhao G, Xiao Y, Zhou X, Dai R, Han M, Wang Z, Xin R, Wang S. Ginsenoside Rd Ameliorates High Fat Diet-Induced Obesity by Enhancing Adaptive Thermogenesis in a cAMP-Dependent Manner. Obesity (Silver Spring) 2020; 28:783-792. [PMID: 32144882 DOI: 10.1002/oby.22761] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/16/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE With the discovery of thermogenic adipocytes in humans, it has been hypothesized that enhancing adaptive thermogenesis may improve obesity. Although many studies have found that ginseng can improve obesity, the beneficial effects of ginsenoside Rd on obesity and its mechanisms have not been studied. METHODS High-fat diet-induced obese mice were used as the study subjects, with intraperitoneal injection of Rd daily at a dose of 15 mg/kg. Body weight and energy metabolism were observed. The effects of Rd on glucose tolerance, insulin sensitivity, and cold tolerance were tested. The expression of genes associated with thermogenesis was analyzed. Finally, the mechanisms by which Rd regulates adaptive thermogenesis were studied. RESULTS Rd ameliorated obesity and insulin resistance. Rd increased cold tolerance through enhancing thermogenic gene expression in brown adipose tissue and increased the browning of white adipose tissue induced by cold stress. Rd increased intracellular cyclic adenosine monophosphate (cAMP) content. Decreasing intracellular cAMP levels by an inhibitor of adenylyl cyclase SQ22536 abolished the promoting effects of Rd on the expression of thermogenic genes. CONCLUSIONS Rd improves obesity and insulin resistance. The upregulation of thermogenesis by Rd is dependent on the cAMP/protein kinase A signaling pathway.
Collapse
Affiliation(s)
- Lu Yao
- College of Basic Medical Sciences, Jilin Medical University, Jilin, China
| | - Zaiqi Han
- College of Pharmacy, Jilin Medical University, Jilin, China
| | - Guoyan Zhao
- Laboratory of Scientific Research, Jilin Medical University, Jilin, China
| | - Yanfang Xiao
- College of Pharmacy, Jilin Medical University, Jilin, China
| | - Xuenan Zhou
- College of Pharmacy, Jilin Medical University, Jilin, China
| | - Rongzheng Dai
- College of Pharmacy, Jilin Medical University, Jilin, China
| | - Meijing Han
- College of Pharmacy, Jilin Medical University, Jilin, China
| | - Zilin Wang
- College of Pharmacy, Jilin Medical University, Jilin, China
| | - Rongshuang Xin
- College of Pharmacy, Jilin Medical University, Jilin, China
| | - Shuran Wang
- College of Public Health, Jilin Medical University, Jilin, China
| |
Collapse
|
45
|
Abdul Sater Z, Jha A, Hamimi A, Mandl A, Hartley IR, Gubbi S, Patel M, Gonzales M, Taïeb D, Civelek AC, Gharib AM, Auh S, O’Mara AE, Pacak K, Cypess AM. Pheochromocytoma and Paraganglioma Patients With Poor Survival Often Show Brown Adipose Tissue Activation. J Clin Endocrinol Metab 2020; 105:5696788. [PMID: 31903484 PMCID: PMC7059996 DOI: 10.1210/clinem/dgz314] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 01/02/2020] [Indexed: 01/22/2023]
Abstract
CONTEXT Pheochromocytomas/paragangliomas (PPGLs) are neuroendocrine tumors that can secrete norepinephrine (NE). Brown adipose tissue (BAT) activation is mediated through the action of NE on β-adrenoceptors (β-ARs). In some malignancies, BAT activation is associated with higher cancer activity. OBJECTIVE To study the relationship between BAT activation and PPGL clinical outcomes. DESIGN A retrospective case-control study that included 342 patients with PPGLs who underwent 18F-fluoro-2-deoxy-D-glucose positron emission tomography-computed tomography (18F-FDG PET/CT) imaging at the National Institutes of Health (NIH). We excluded all patients with parasympathetic tumors and those who underwent 18F-FDG PET/CT after PPGL resection. Scans of 205 patients were reviewed by 2 blinded nuclear medicine physicians; 16 patients had BAT activation on 18F-FDG PET/CT [7.80%; age 27.50 (15.00-45.50) years; 10 female/6 male; body mass index [BMI] 24.90 [19.60-25.35] kg/m2). From the remaining 189 patients, we selected 36 matched controls (age 34.4 [25.4-45.5] years; 21 female/15 male; BMI 25.0 [22.0-26.0] kg/m2). PRIMARY OUTCOME MEASURE Overall survival. RESULTS The presence of active BAT on 18F-FDG PET/CT was associated with decreased overall survival when compared with the control group (HRz 5.80; 95% CI, 1.05-32.05; P = 0.02). This association remained significant after adjusting for the SDHB mutation. Median plasma NE in the BAT group was higher than the control group [4.65 vs 0.55 times above the upper limit of normal; P < 0.01]. There was a significant association between higher plasma NE levels and mortality in PPGLs in both groups. CONCLUSIONS Our findings suggest that the detection of BAT activity in PPGL patients is associated with higher mortality. We suggest that BAT activation could either be reflecting or contributing to a state of increased host stress that may predict poor outcome in metastatic PPGL.
Collapse
Affiliation(s)
- Zahraa Abdul Sater
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Abhishek Jha
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Ahmed Hamimi
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Adel Mandl
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Iris R Hartley
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Sriram Gubbi
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Mayank Patel
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Melissa Gonzales
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Ali Cahid Civelek
- Nuclear Medicine Division, Radiology, and Imaging Sciences, Warren Grant Magnuson Clinical Center, National Institutes of Health, Bethesda, Maryland
- Nuclear Medicine, Radiology and Radiological Science, Johns Hopkins Medicine, Baltimore, Maryland
| | - Ahmed M Gharib
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Sungyoung Auh
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Alana E O’Mara
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Aaron M Cypess
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
- Correspondence and Reprint Requests: Aaron M. Cypess, MD, PhD, MMSc, Acting Section Chief, Translational Physiology Section, Diabetes, Endocrinology, and Obesity Branch, 10 Center Drive, Building 10, CRC, Room, Bethesda, Maryland 20892–1109 USA. E-mail:
| |
Collapse
|
46
|
Abstract
Animals that lack the hormone leptin become grossly obese, purportedly for 2 reasons: increased food intake and decreased energy expenditure (thermogenesis). This review examines the experimental evidence for the thermogenesis component. Analysis of the data available led us to conclude that the reports indicating hypometabolism in the leptin-deficient ob/ob mice (as well as in the leptin-receptor-deficient db/db mice and fa/fa rats) derive from a misleading calculation artefact resulting from expression of energy expenditure per gram of body weight and not per intact organism. Correspondingly, the body weight-reducing effects of leptin are not augmented by enhanced thermogenesis. Congruent with this, there is no evidence that the ob/ob mouse demonstrates atrophied brown adipose tissue or diminished levels of total UCP1 mRNA or protein when the ob mutation is studied on the inbred C57BL/6 mouse background, but a reduced sympathetic nerve activity is observed. On the outbred "Aston" mouse background, brown adipose tissue atrophy is seen, but whether this is of quantitative significance for the development of obesity has not been demonstrated. We conclude that leptin is not a thermogenic hormone. Rather, leptin has effects on body temperature regulation, by opposing torpor bouts and by shifting thermoregulatory thresholds. The central pathways behind these effects are largely unexplored.
Collapse
Affiliation(s)
- Alexander W Fischer
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, Stockholm, Sweden.,Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, Stockholm, Sweden
| |
Collapse
|
47
|
Adeghate E, Lotfy M, D'Souza C, Alseiari SM, Alsaadi AA, Qahtan SA. Hypocretin/orexin modulates body weight and the metabolism of glucose and insulin. Diabetes Metab Res Rev 2020; 36:e3229. [PMID: 31655012 DOI: 10.1002/dmrr.3229] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 08/16/2019] [Accepted: 10/06/2019] [Indexed: 12/18/2022]
Abstract
The hypocretin/orexin (Hcrt/orexin) unit affects the functions of the nervous, cardiovascular, gastrointestinal, and reproductive systems. Hcrt/orexin ligands and receptors have been localized to different parts of the central and peripheral nervous systems, cerebrospinal fluid and blood, exocrine (pancreas, salivary, lacrimal) as well as endocrine (pancreatic islets, pituitary, adrenal) glands. Several factors including stress, glucagon-like peptide-1 agonists, glutamate, nicotine, glucose, and hypoglycaemia stimulate the expression of Hcrt/orexin system, but it is inhibited by ageing, bone morphogenetic protein, hypoxia/hypercapnia, melanocortin receptor accessory protein 2, and glucagon. Literature reports show that Hcrt/orexin can significantly increase insulin secretion from normal and diabetic rat pancreata. Hcrt/orexin decreases blood glucose concentration and reduces insulin resistance partly via increased tissue expression of glucose transporter type 4. It reduces obesity by increasing browning of fat cells and energy expenditure. Taken together, Hcrt/orexin modulates obesity and the metabolism of glucose and insulin. The Hcrt/orexin system may thus be a target in the development of new therapies for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Ernest Adeghate
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohamed Lotfy
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Crystal D'Souza
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Saleh Meqbel Alseiari
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Abdulla Ali Alsaadi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Saif Abdo Qahtan
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
48
|
Xia B, Shi XC, Xie BC, Zhu MQ, Chen Y, Chu XY, Cai GH, Liu M, Yang SZ, Mitchell GA, Pang WJ, Wu JW. Urolithin A exerts antiobesity effects through enhancing adipose tissue thermogenesis in mice. PLoS Biol 2020; 18:e3000688. [PMID: 32218572 PMCID: PMC7141696 DOI: 10.1371/journal.pbio.3000688] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 04/08/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity leads to multiple health problems, including diabetes, fatty liver, and even cancer. Here, we report that urolithin A (UA), a gut-microflora-derived metabolite of pomegranate ellagitannins (ETs), prevents diet-induced obesity and metabolic dysfunctions in mice without causing adverse effects. UA treatment increases energy expenditure (EE) by enhancing thermogenesis in brown adipose tissue (BAT) and inducing browning of white adipose tissue (WAT). Mechanistically, UA-mediated increased thermogenesis is caused by an elevation of triiodothyronine (T3) levels in BAT and inguinal fat depots. This is also confirmed in UA-treated white and brown adipocytes. Consistent with this mechanism, UA loses its beneficial effects on activation of BAT, browning of white fat, body weight control, and glucose homeostasis when thyroid hormone (TH) production is blocked by its inhibitor, propylthiouracil (PTU). Conversely, administration of exogenous tetraiodothyronine (T4) to PTU-treated mice restores UA-induced activation of BAT and browning of white fat and its preventive role on high-fat diet (HFD)-induced weight gain. Together, these results suggest that UA is a potent antiobesity agent with potential for human clinical applications.
Collapse
Affiliation(s)
- Bo Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiao Chen Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Bao Cai Xie
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Meng Qing Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yan Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xin Yi Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Guo He Cai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Min Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Shi Zhen Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Grant A. Mitchell
- Division of Medical Genetics, Department of Paediatrics, Université de Montréal and Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec, Canada
| | - Wei Jun Pang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jiang Wei Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
49
|
Seoane-Collazo P, Martínez-Sánchez N, Milbank E, Contreras C. Incendiary Leptin. Nutrients 2020; 12:nu12020472. [PMID: 32069871 PMCID: PMC7071158 DOI: 10.3390/nu12020472] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/06/2020] [Accepted: 02/08/2020] [Indexed: 02/08/2023] Open
Abstract
Leptin is a hormone released by adipose tissue that plays a key role in the control of energy homeostasis through its binding to leptin receptors (LepR), mainly expressed in the hypothalamus. Most scientific evidence points to leptin’s satiating effect being due to its dual capacity to promote the expression of anorexigenic neuropeptides and to reduce orexigenic expression in the hypothalamus. However, it has also been demonstrated that leptin can stimulate (i) thermogenesis in brown adipose tissue (BAT) and (ii) the browning of white adipose tissue (WAT). Since the demonstration of the importance of BAT in humans 10 years ago, its study has aroused great interest, mainly in the improvement of obesity-associated metabolic disorders through the induction of thermogenesis. Consequently, several strategies targeting BAT activation (mainly in rodent models) have demonstrated great potential to improve hyperlipidemias, hepatic steatosis, insulin resistance and weight gain, leading to an overall healthier metabolic profile. Here, we review the potential therapeutic ability of leptin to correct obesity and other metabolic disorders, not only through its satiating effect, but by also utilizing its thermogenic properties.
Collapse
Affiliation(s)
- Patricia Seoane-Collazo
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| | - Noelia Martínez-Sánchez
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| | - Edward Milbank
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Cristina Contreras
- Department of Physiology, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| |
Collapse
|
50
|
Bastías-Pérez M, Zagmutt S, Soler-Vázquez MC, Serra D, Mera P, Herrero L. Impact of Adaptive Thermogenesis in Mice on the Treatment of Obesity. Cells 2020; 9:E316. [PMID: 32012991 PMCID: PMC7072509 DOI: 10.3390/cells9020316] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/20/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity and associated metabolic diseases have become a priority area of study due to the exponential increase in their prevalence and the corresponding health and economic impact. In the last decade, brown adipose tissue has become an attractive target to treat obesity. However, environmental variables such as temperature and the dynamics of energy expenditure could influence brown adipose tissue activity. Currently, most metabolic studies are carried out at a room temperature of 21 °C, which is considered a thermoneutral zone for adult humans. However, in mice this chronic cold temperature triggers an increase in their adaptive thermogenesis. In this review, we aim to cover important aspects related to the adaptation of animals to room temperature, the influence of housing and temperature on the development of metabolic phenotypes in experimental mice and their translation to human physiology. Mice studies performed in chronic cold or thermoneutral conditions allow us to better understand underlying physiological mechanisms for successful, reproducible translation into humans in the fight against obesity and metabolic diseases.
Collapse
Affiliation(s)
- Marianela Bastías-Pérez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - Sebastián Zagmutt
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - M Carmen Soler-Vázquez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Paula Mera
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|