1
|
Jeon YG, Kim SW, Kim JB. Decoding temporal thermogenesis: coregulator selectivity and transcriptional control in brown and beige adipocytes. Adipocyte 2024; 13:2391511. [PMID: 39155481 PMCID: PMC11340756 DOI: 10.1080/21623945.2024.2391511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024] Open
Abstract
In mammals, brown adipose tissue (BAT) and beige adipocytes in white adipose tissue (WAT) play pivotal roles in maintaining body temperature and energy metabolism. In mice, BAT quickly stimulates thermogenesis by activating brown adipocytes upon cold exposure. In the presence of chronic cold stimuli, beige adipocytes are recruited in inguinal WAT to support heat generation. Accumulated evidence has shown that thermogenic execution of brown and beige adipocytes is regulated in a fat depot-specific manner. Recently, we have demonstrated that ubiquitin ligase ring finger protein 20 (RNF20) regulates brown and beige adipocyte thermogenesis through fat-depot-specific modulation. In BAT, RNF20 regulates transcription factor GA-binding protein alpha (GABPα), whereas in inguinal WAT, RNF20 potentiates transcriptional activity of peroxisome proliferator-activated receptor-gamma (PPARγ) through the degradation of nuclear corepressor 1 (NCoR1). This study proposes the molecular mechanisms by which co-regulator(s) selectively and temporally control transcription factors to coordinate adipose thermogenesis in a fat-depot-specific manner. In this Commentary, we provide molecular features of brown and beige adipocyte thermogenesis and discuss the underlying mechanisms of distinct thermogenic processes in two fat depots.
Collapse
Affiliation(s)
- Yong Geun Jeon
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Sun Won Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jae Bum Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
2
|
Sun DD, Li XQ, Liu YT, Ge MQ, Hou ZC. The Application of Duck Embryonic Fibroblasts CCL-141 as a Cell Model for Adipogenesis. Animals (Basel) 2024; 14:2973. [PMID: 39457903 PMCID: PMC11503743 DOI: 10.3390/ani14202973] [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: 08/24/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
The duck embryo fibroblast cell line CCL-141, which is currently the only commercialized duck cell line, has been underexplored in adipogenesis research. (1) Background: This study establishes an experimental protocol to induce adipogenesis in CCL-141 cells, addressing the importance of understanding gene functions in this process. (2) Methods: Chicken serum, fatty acids, insulin, and all-trans retinoic acid were used to treat CCL-141 cells, with adipogenesis confirmed by Oil Red O staining and gene expression quantification. CRISPR/Cas9 technology was applied to knockout PPARγ, and the resulting adipogenic phenotype was assessed. (3) Results: The treatments promoted adipogenesis, and the knockout of PPARγ validated the cell line's utility for gene function studies. (4) Conclusions: CCL-141 cells are a suitable model for investigating duck adipogenesis, contributing to the understanding of regulatory factors in this biological process.
Collapse
Affiliation(s)
- Dan-Dan Sun
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (D.-D.S.); (X.-Q.L.); (Y.-T.L.); (M.-Q.G.)
| | - Xiao-Qin Li
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (D.-D.S.); (X.-Q.L.); (Y.-T.L.); (M.-Q.G.)
| | - Yong-Tong Liu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (D.-D.S.); (X.-Q.L.); (Y.-T.L.); (M.-Q.G.)
| | - Meng-Qi Ge
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (D.-D.S.); (X.-Q.L.); (Y.-T.L.); (M.-Q.G.)
| | - Zhuo-Cheng Hou
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (D.-D.S.); (X.-Q.L.); (Y.-T.L.); (M.-Q.G.)
| |
Collapse
|
3
|
Mo YY, Han YX, Xu SN, Jiang HL, Wu HX, Cai JM, Li L, Bu YH, Xiao F, Liang HD, Wen Y, Liu YZ, Yin YL, Zhou HD. Adipose Tissue Plasticity: A Comprehensive Definition and Multidimensional Insight. Biomolecules 2024; 14:1223. [PMID: 39456156 PMCID: PMC11505740 DOI: 10.3390/biom14101223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Adipose tissue is composed of adipocytes, stromal vascular fraction, nerves, surrounding immune cells, and the extracellular matrix. Under various physiological or pathological conditions, adipose tissue shifts cellular composition, lipid storage, and organelle dynamics to respond to the stress; this remodeling is called "adipose tissue plasticity". Adipose tissue plasticity includes changes in the size, species, number, lipid storage capacity, and differentiation function of adipocytes, as well as alterations in the distribution and cellular composition of adipose tissue. This plasticity has a major role in growth, obesity, organismal protection, and internal environmental homeostasis. Moreover, certain thresholds exist for this plasticity with significant individualized differences. Here, we comprehensively elaborate on the specific connotation of adipose tissue plasticity and the relationship between this plasticity and the development of many diseases. Meanwhile, we summarize possible strategies for treating obesity in response to adipose tissue plasticity, intending to provide new insights into the dynamic changes in adipose tissue and contribute new ideas to relevant clinical problems.
Collapse
Affiliation(s)
- Yu-Yao Mo
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Xin Han
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Shi-Na Xu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hong-Li Jiang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hui-Xuan Wu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Jun-Min Cai
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Long Li
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yan-Hong Bu
- Department of Blood Transfusion, The Second Xiangya Hospital, Central South University, Changsha 410012, China;
| | - Fen Xiao
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Han-Dan Liang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Ying Wen
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Ze Liu
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China;
| | - Yu-Long Yin
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| |
Collapse
|
4
|
Wang R, Gomez Salazar M, Pruñonosa Cervera I, Coutts A, French K, Pinto MM, Gohlke S, García-Martín R, Blüher M, Schofield CJ, Kourtzelis I, Stimson RH, Bénézech C, Christian M, Schulz TJ, Gudmundsson EF, Jennings LL, Gudnason VG, Chavakis T, Morton NM, Emilsson V, Michailidou Z. Adipocyte deletion of the oxygen-sensor PHD2 sustains elevated energy expenditure at thermoneutrality. Nat Commun 2024; 15:7483. [PMID: 39209825 PMCID: PMC11362468 DOI: 10.1038/s41467-024-51718-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Enhancing thermogenic brown adipose tissue (BAT) function is a promising therapeutic strategy for metabolic disease. However, predominantly thermoneutral modern human living conditions deactivate BAT. We demonstrate that selective adipocyte deficiency of the oxygen-sensor HIF-prolyl hydroxylase (PHD2) gene overcomes BAT dormancy at thermoneutrality. Adipocyte-PHD2-deficient mice maintain higher energy expenditure having greater BAT thermogenic capacity. In human and murine adipocytes, a PHD inhibitor increases Ucp1 levels. In murine brown adipocytes, antagonising the major PHD2 target, hypoxia-inducible factor-(HIF)-2a abolishes Ucp1 that cannot be rescued by PHD inhibition. Mechanistically, PHD2 deficiency leads to HIF2 stabilisation and binding of HIF2 to the Ucp1 promoter, thus enhancing its expression in brown adipocytes. Serum proteomics analysis of 5457 participants in the deeply phenotyped Age, Gene and Environment Study reveal that serum PHD2 associates with increased risk of metabolic disease. Here we show that adipose-PHD2-inhibition is a therapeutic strategy for metabolic disease and identify serum PHD2 as a disease biomarker.
Collapse
Affiliation(s)
- Rongling Wang
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mario Gomez Salazar
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Iris Pruñonosa Cervera
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Amanda Coutts
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Karen French
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Marlene Magalhaes Pinto
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Sabrina Gohlke
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Ruben García-Martín
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC (CNB-CSIC), Campus-UAM, Madrid, Spain
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research University of Oxford, Oxford, UK
| | - Ioannis Kourtzelis
- Hull York Medical School, York Biomedical Research Institute, University of York, York, UK
| | - Roland H Stimson
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mark Christian
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | | | - Lori L Jennings
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Vilmundur G Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Triantafyllos Chavakis
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Nicholas M Morton
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Valur Emilsson
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Zoi Michailidou
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK.
| |
Collapse
|
5
|
Sharma AK, Khandelwal R, Wolfrum C. Futile cycles: Emerging utility from apparent futility. Cell Metab 2024; 36:1184-1203. [PMID: 38565147 DOI: 10.1016/j.cmet.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/15/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Futile cycles are biological phenomena where two opposing biochemical reactions run simultaneously, resulting in a net energy loss without appreciable productivity. Such a state was presumed to be a biological aberration and thus deemed an energy-wasting "futile" cycle. However, multiple pieces of evidence suggest that biological utilities emerge from futile cycles. A few established functions of futile cycles are to control metabolic sensitivity, modulate energy homeostasis, and drive adaptive thermogenesis. Yet, the physiological regulation, implication, and pathological relevance of most futile cycles remain poorly studied. In this review, we highlight the abundance and versatility of futile cycles and propose a classification scheme. We further discuss the energetic implications of various futile cycles and their impact on basal metabolic rate, their bona fide and tentative pathophysiological implications, and putative drug interactions.
Collapse
Affiliation(s)
- Anand Kumar Sharma
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Radhika Khandelwal
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
6
|
Torghabeh FD, Javadi B, Sahebkar A. Dietary anethole: a systematic review of its protective effects against metabolic syndrome. J Diabetes Metab Disord 2024; 23:619-631. [PMID: 38932801 PMCID: PMC11196516 DOI: 10.1007/s40200-023-01322-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/23/2023] [Indexed: 06/28/2024]
Abstract
Background Metabolic syndrome (MetS) is a cluster of physiological, biochemical, clinical, and metabolic conditions that aggravate the risk of severe diseases such as cardiovascular disease, type 2 diabetes mellitus, and fatty liver. Several dietary molecules have been considered preventive compounds for MetS. Anethole, a natural phenylpropanoid, has been found to protect against MetS and its associated components. Aim This systematic review aims to provide an overview of the preclinical evidence supporting the protective effects of dietary anethole against MetS and the associated diseases. Methods A literature search was performed using Web of Sciences, PubMed, Scopus, and Google Scholar to identify studies reporting the protective effects of dietary anethole against MetS, without any time restrictions. Review articles, letters to editors, editorials, unpublished results, and non-English papers were excluded from the study. Results The results showed that anethole has the potential to effectively protect against the key features of MetS via various mechanisms, including antioxidant and anti-inflammatory effects, stimulating insulin secretion from β-cells, mediating oxidative stress, modulation of the mTOR/PPARγ axis, arterial remodeling, and improvement of vascular relaxation. Conclusion Anethole modulates several molecular pathways that are implicated in the pathogenesis of MetS. Future in vitro and animal investigations should be conducted to explore other anti-MetS signaling pathways of anethole. Additionally, well-designed clinical studies are warranted to determine the optimal human dose, bioavailability, and pharmacokinetic characteristics of this dietary compound.
Collapse
Affiliation(s)
| | - Behjat Javadi
- Department of Traditional Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Winn NC, Schleh MW, Garcia JN, Lantier L, McGuinness OP, Blair JA, Hasty AH, Wasserman DH. Insulin at the intersection of thermoregulation and glucose homeostasis. Mol Metab 2024; 81:101901. [PMID: 38354854 PMCID: PMC10877958 DOI: 10.1016/j.molmet.2024.101901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/16/2024] Open
Abstract
Mammals are protected from changes in environmental temperature by altering energetic processes that modify heat production. Insulin is the dominant stimulus of glucose uptake and metabolism, which are fundamental for thermogenic processes. The purpose of this work was to determine the interaction of ambient temperature induced changes in energy expenditure (EE) on the insulin sensitivity of glucose fluxes. Short-term and adaptive responses to thermoneutral temperature (TN, ∼28 °C) and room (laboratory) temperature (RT, ∼22 °C) were studied in mice. This range of temperature does not cause detectable changes in circulating catecholamines or shivering and postabsorptive glucose homeostasis is maintained. We tested the hypothesis that a decrease in EE that occurs with TN causes insulin resistance and that this reduction in insulin action and EE is reversed upon short term (<12h) transition to RT. Insulin-stimulated glucose disposal (Rd) and tissue-specific glucose metabolic index were assessed combining isotopic tracers with hyperinsulinemic-euglycemic clamps. EE and insulin-stimulated Rd are both decreased (∼50%) in TN-adapted vs RT-adapted mice. When RT-adapted mice are switched to TN, EE rapidly decreases and Rd is reduced by ∼50%. TN-adapted mice switched to RT exhibit a rapid increase in EE, but whole-body insulin-stimulated Rd remains at the low rates of TN-adapted mice. In contrast, whole body glycolytic flux rose with EE. This higher EE occurs without increasing glucose uptake from the blood, but rather by diverting glucose from glucose storage to glycolysis. In addition to adaptations in insulin action, 'insulin-independent' glucose uptake in brown fat is exquisitely sensitive to thermoregulation. These results show that insulin action adjusts to non-stressful changes in ambient temperature to contribute to the support of body temperature homeostasis without compromising glucose homeostasis.
Collapse
Affiliation(s)
- Nathan C Winn
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| | - Michael W Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jamie N Garcia
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - Joslin A Blair
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| |
Collapse
|
8
|
Hsu CY, Liao CC, Lin ZC, Alalaiwe A, Hwang E, Lin TW, Fang JY. Facile adipocyte uptake and liver/adipose tissue delivery of conjugated linoleic acid-loaded tocol nanocarriers for a synergistic anti-adipogenesis effect. J Nanobiotechnology 2024; 22:50. [PMID: 38317220 PMCID: PMC10845550 DOI: 10.1186/s12951-024-02316-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/26/2024] [Indexed: 02/07/2024] Open
Abstract
Obesity is a major risk to human health. Adipogenesis is blocked by α-tocopherol and conjugated linoleic acid (CLA). However, their effect at preventing obesity is uncertain. The effectiveness of the bioactive agents is associated with their delivery method. Herein, we designed CLA-loaded tocol nanostructured lipid carriers (NLCs) for enhancing the anti-adipogenic activity of α-tocopherol and CLA. Adipogenesis inhibition by the nanocarriers was examined using an in vitro adipocyte model and an in vivo rat model fed a high fat diet (HFD). The targeting of the tocol NLCs into adipocytes and adipose tissues were also investigated. A synergistic anti-adipogenesis effect was observed for the combination of free α-tocopherol and CLA. Nanoparticles with different amounts of solid lipid were developed with an average size of 121‒151 nm. The NLCs with the smallest size (121 nm) showed greater adipocyte internalization and differentiation prevention than the larger size. The small-sized NLCs promoted CLA delivery into adipocytes by 5.5-fold as compared to free control. The nanocarriers reduced fat accumulation in adipocytes by counteracting the expression of the adipogenic transcription factors peroxisome proliferator activated receptor (PPAR)γ and CCAAT/enhancer-binding protein (C/EBP)α, and lipogenic enzymes acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS). Localized administration of CLA-loaded tocol NLCs significantly reduced body weight, total cholesterol, and liver damage indicators in obese rats. The biodistribution study demonstrated that the nanoparticles mainly accumulated in liver and adipose tissues. The NLCs decreased adipocyte hypertrophy and cytokine overexpression in the groin and epididymis to a greater degree than the combination of free α-tocopherol and CLA. In conclusion, the lipid-based nanocarriers were verified to inhibit adipogenesis in an efficient and safe way.
Collapse
Affiliation(s)
- Ching-Yun Hsu
- Department of Nutrition and Health Sciences, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan
| | - Chia-Chih Liao
- Department of Anesthesiology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Zih-Chan Lin
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi, Chiayi, Taiwan
| | - Ahmed Alalaiwe
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Erica Hwang
- Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tzu-Wei Lin
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, 259 Wen-Hwa 1st Road, Kweishan, Taoyuan, 333, Taiwan
| | - Jia-You Fang
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan.
- Department of Anesthesiology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, 259 Wen-Hwa 1st Road, Kweishan, Taoyuan, 333, Taiwan.
| |
Collapse
|
9
|
Lee HS, Choi SM, Lim SH, Choi CI. Betanin from Beetroot ( Beta vulgaris L.) Regulates Lipid Metabolism and Promotes Fat Browning in 3T3-L1 Adipocytes. Pharmaceuticals (Basel) 2023; 16:1727. [PMID: 38139853 PMCID: PMC10748323 DOI: 10.3390/ph16121727] [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: 11/08/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Fat browning, which converts white adipose tissue to brown, has attracted attention as a promising strategy for the treatment of obesity. Betanin (BT) has been reported to have potential anti-obesity activity. 3T3-L1 cells were differentiated for 7 days during BT treatment. The BT concentration range for the study was determined using an MTT assay, and lipid accumulation was evaluated by Oil-Red-O staining. The expression of protein level was analyzed by Western blot. Immunofluorescence images were performed with confocal microscopy to visually show the amount and location of thermogenesis factor uncoupling protein1 (UCP1) and mitochondria. qRT-PCR was performed to evaluate mRNA expression. BT inhibited lipid accumulation and increased the expression of UCP1, peroxisome-proliferator-activated receptor gamma (PPARγ), and PPARγ coactivator-1 alpha (PGC-1α). In addition, the increases in beige adipocyte-specific markers were observed, supporting BT-mediated browning of the fat tissue. The UCP1 was localized in the inner membrane of the mitochondria, and its expression was associated with mitochondrial activation. Consistent with this, the mRNA expression of mitochondrial biogenesis markers increased in 3T3-L1 cells after BT treatment. Immunofluorescence staining also indicated an increased number of mitochondria and UCP1, respectively. Moreover, BT inhibited lipogenesis and enhanced lipolysis and fatty acid oxidation. This mechanism has been suggested to be mediated by an adenosine monophosphate-activated protein kinase (AMPK) pathway. BT induces fat browning and regulates lipid metabolism via the AMPK-mediated pathway in 3T3-L1 cells, suggesting that BT can be a promising candidate for controlling obesity.
Collapse
Affiliation(s)
| | | | | | - Chang-Ik Choi
- Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (H.S.L.); (S.M.C.); (S.H.L.)
| |
Collapse
|
10
|
Winn NC, Schleh MW, Garcia JN, Lantier L, McGuinness OP, Blair JA, Hasty AH, Wasserman DH. Insulin at the Intersection of Thermoregulation and Glucose Homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.566254. [PMID: 38014310 PMCID: PMC10680846 DOI: 10.1101/2023.11.17.566254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Mammals are protected from changes in environmental temperature by altering energetic processes that modify heat production. Insulin is the dominant stimulus of glucose uptake and metabolism, which are fundamental for thermogenic processes. The purpose of this work was to determine the interaction of ambient temperature induced changes in energy expenditure (EE) on the insulin sensitivity of glucose fluxes. Short-term and adaptive responses to thermoneutral temperature (TN, ~28°C) and room (laboratory) temperature (RT, ~22°C) were studied in mice. This range of temperature does not cause detectable changes in circulating catecholamines or shivering and postabsorptive glucose homeostasis is maintained. We tested the hypothesis that a decrease in EE that occurs with TN causes insulin resistance and that this reduction in insulin action and EE is reversed upon short term (<12h) transition to RT. Insulin-stimulated glucose disposal (Rd) and tissue specific glucose uptake were assessed combining isotopic tracers with hyperinsulinemic-euglycemic clamps. EE and insulin-stimulated Rd are both decreased (~50%) in TN-adapted vs RT-adapted mice. When RT-adapted mice are switched to TN, EE rapidly decreases and Rd is reduced by ~50%. TN-adapted mice switched to RT exhibit a rapid increase in EE, but whole body insulin-stimulated Rd remains at the low rates of TN-adapted mice. In contrast, whole body glycolytic flux rose with EE. This higher EE occurs without increasing glucose uptake from the blood, but rather by diverting glucose from glucose storage to glycolysis. In addition to adaptations in insulin action, 'insulin-independent' glucose uptake in brown fat is exquisitely sensitive to thermoregulation. These results show that insulin action adjusts to non-stressful changes in ambient temperature to contribute to the support of body temperature homeostasis without compromising glucose homeostasis.
Collapse
Affiliation(s)
- Nathan C. Winn
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Michael W. Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Jamie N. Garcia
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, Tennessee, USA
| | - Owen P. McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, Tennessee, USA
| | - Joslin A. Blair
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Alyssa H. Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - David H. Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, Tennessee, USA
| |
Collapse
|
11
|
Tang Y, Wang YD, Wang YY, Liao ZZ, Xiao XH. Skeletal muscles and gut microbiota-derived metabolites: novel modulators of adipocyte thermogenesis. Front Endocrinol (Lausanne) 2023; 14:1265175. [PMID: 37867516 PMCID: PMC10588486 DOI: 10.3389/fendo.2023.1265175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Obesity occurs when overall energy intake surpasses energy expenditure. White adipose tissue is an energy storage site, whereas brown and beige adipose tissues catabolize stored energy to generate heat, which protects against obesity and obesity-associated metabolic disorders. Metabolites are substrates in metabolic reactions that act as signaling molecules, mediating communication between metabolic sites (i.e., adipose tissue, skeletal muscle, and gut microbiota). Although the effects of metabolites from peripheral organs on adipose tissue have been extensively studied, their role in regulating adipocyte thermogenesis requires further investigation. Skeletal muscles and intestinal microorganisms are important metabolic sites in the body, and their metabolites play an important role in obesity. In this review, we consolidated the latest research on skeletal muscles and gut microbiota-derived metabolites that potentially promote adipocyte thermogenesis. Skeletal muscles can release lactate, kynurenic acid, inosine, and β-aminoisobutyric acid, whereas the gut secretes bile acids, butyrate, succinate, cinnabarinic acid, urolithin A, and asparagine. These metabolites function as signaling molecules by interacting with membrane receptors or controlling intracellular enzyme activity. The mechanisms underlying the reciprocal exchange of metabolites between the adipose tissue and other metabolic organs will be a focal point in future studies on obesity. Furthermore, understanding how metabolites regulate adipocyte thermogenesis will provide a basis for establishing new therapeutic targets for obesity.
Collapse
Affiliation(s)
- Yi Tang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ya-Di Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan-Yuan Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhe-Zhen Liao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin-Hua Xiao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
12
|
Hong J, Shi Y, Chen J, Mi M, Ren Q, Zhang Y, Shen M, Bu J, Kang Y. Konjac glucomannan attenuate high-fat diet-fed obesity through enhancing β-adrenergic-mediated thermogenesis in inguinal white adipose tissue in mice. Glycoconj J 2023; 40:575-586. [PMID: 37535173 DOI: 10.1007/s10719-023-10131-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: 09/27/2022] [Revised: 06/28/2023] [Accepted: 07/19/2023] [Indexed: 08/04/2023]
Abstract
Konjac glucomannan (KGM) has been reported to prevent high-fat diet-induced obesity, and we study investigated whether dietary supplementation with KGM can prevent obesity by increasing energy expenditure in inguinal white adipose tissue (iWAT) of high-fat diet (HF) -fed mice. Weaned mice fed the control diet (Con), HF, or HF plus KGM (8%, w/w, HFK) were divided into three groups. The results showed that 10-week supplementation with KGM significantly reduced partial adipose tissue weight and body weight, and improved glucose tolerance. Compared to the HF group, plasma lipid concentrations in the HFK group were greatly decreased to the control level. Moreover, transcriptomic research has shown that genes that are mainly associated with energy and lipid metabolism are significantly altered in iWAT. Mechanistically, KGM stimulated thermogenesis by promoting the expression of uncoupling protein-1 (UCP1) and the β3-adrenergic receptor (ADR3β). Taken together, our results suggest that dietary supplementation with konjac glucomannan can effectively alleviate obesity induced by a high-fat diet by activating ADR3β-mediated iWAT thermogenesis. Dietary supplementation with KGM can effectively alleviate high fat diet- induced obesity mice by via activating ADR3β-mediated thermogenesis of iWAT.
Collapse
Affiliation(s)
- Jian Hong
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Yancheng Teachers University, Yancheng, 224007, Jiangsu, China
- College of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng, 224051, Jiangsu, China
| | - Yun Shi
- College of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng, 224051, Jiangsu, China
- Tibetan Traditional Medicine College, Lhasa, 850000, Xizang, China
| | - Jing Chen
- College of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng, 224051, Jiangsu, China
| | - Ma Mi
- College of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng, 224051, Jiangsu, China
| | - Qingjia Ren
- College of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng, 224051, Jiangsu, China
| | - Yanzhou Zhang
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Yancheng Teachers University, Yancheng, 224007, Jiangsu, China
| | - Min Shen
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Yancheng Teachers University, Yancheng, 224007, Jiangsu, China
| | - Jing Bu
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Yancheng Teachers University, Yancheng, 224007, Jiangsu, China
| | - Yijun Kang
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Yancheng Teachers University, Yancheng, 224007, Jiangsu, China.
| |
Collapse
|
13
|
Gupta A, Efthymiou V, Kodani SD, Shamsi F, Patti ME, Tseng YH, Streets A. Mapping the transcriptional landscape of human white and brown adipogenesis using single-nuclei RNA-seq. Mol Metab 2023; 74:101746. [PMID: 37286033 PMCID: PMC10338377 DOI: 10.1016/j.molmet.2023.101746] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023] Open
Abstract
Adipogenesis is key to maintaining organism-wide energy balance and healthy metabolic phenotype, making it critical to thoroughly comprehend its molecular regulation in humans. By single-nuclei RNA-sequencing (snRNA-seq) of over 20,000 differentiating white and brown preadipocytes, we constructed a high-resolution temporal transcriptional landscape of human white and brown adipogenesis. White and brown preadipocytes were isolated from a single individual's neck region, thereby eliminating inter-subject variability across two distinct lineages. These preadipocytes were also immortalized to allow for controlled, in vitro differentiation, allowing sampling of distinct cellular states across the spectrum of adipogenic progression. Pseudotemporal cellular ordering revealed the dynamics of ECM remodeling during early adipogenesis, and lipogenic/thermogenic response during late white/brown adipogenesis. Comparison with adipogenic regulation in murine models Identified several novel transcription factors as potential targets for adipogenic/thermogenic drivers in humans. Among these novel candidates, we explored the role of TRPS1 in adipocyte differentiation and showed that its knockdown impairs white adipogenesis in vitro. Key adipogenic and lipogenic markers revealed in our analysis were applied to analyze publicly available scRNA-seq datasets; these confirmed unique cell maturation features in recently discovered murine preadipocytes, and revealed inhibition of adipogenic expansion in humans with obesity. Overall, our study presents a comprehensive molecular description of both white and brown adipogenesis in humans and provides an important resource for future studies of adipose tissue development and function in both health and metabolic disease state.
Collapse
Affiliation(s)
- Anushka Gupta
- University of California at Berkeley, University of California at San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
| | - Vissarion Efthymiou
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Sean D Kodani
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Farnaz Shamsi
- Department of Molecular Pathobiology, New York University, New York, NY 10010, USA
| | - Mary Elizabeth Patti
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Yu-Hua Tseng
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Aaron Streets
- University of California at Berkeley, University of California at San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA; Biophysics Graduate Group, University of California at Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
14
|
Bagon BB, Lee J, Matienzo ME, Lee SJ, Pak SW, Kim K, Lee J, Lee CM, Shin IS, Moon C, Park MJ, Kim DI. Cold-induced adaptive thermogenesis is impaired by exposure of Asian sand dust in mice. J Therm Biol 2023; 116:103675. [PMID: 37517326 DOI: 10.1016/j.jtherbio.2023.103675] [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: 01/11/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 08/01/2023]
Abstract
Desertification and desert sandstorms caused by the worsening global warming pose increasing risks to human health. In particular, Asian sand dust (ASD) exposure has been related to an increase in mortality and hospital admissions for respiratory diseases. In this study, we investigated the effects of ASD on metabolic tissues in comparison to diesel particulate matter (DPM) that is known to cause adverse health effects. We found that larger lipid droplets were accumulated in the brown adipose tissues (BAT) of ASD-administered but not DPM-administered mice. Thermogenic gene expression was decreased in these mice as well. When ASD-administered mice were exposed to the cold, they failed to maintain their body temperature, suggesting that the ASD administration had led to impairments in cold-induced adaptive thermogenesis. However, impaired thermogenesis was not observed in DPM-administered mice. Furthermore, mice fed a high-fat diet that were chronically administered ASD demonstrated unexplained weight loss, indicating that chronic administration of ASD could be lethal in obese mice. We further identified that ASD-induced lung inflammation was not exacerbated in uncoupling protein 1 knockout mice, whose thermogenic capacity is impaired. Collectively, ASD exposure can impair cold-induced adaptive thermogenic responses in mice and increase the risk of mortality in obese mice.
Collapse
Affiliation(s)
- Bernadette B Bagon
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Junhyeong Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Merc Emil Matienzo
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Se-Jin Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea; Department of Veterinary Pharmacology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - So-Won Pak
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea; Department of Veterinary Pharmacology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Keon Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea; Department of Veterinary Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Jeongmin Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea; Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Chang-Min Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea; Department of Veterinary Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - In-Sik Shin
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea; Department of Veterinary Pharmacology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Changjong Moon
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea; Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Min-Jung Park
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea.
| | - Dong-Il Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea.
| |
Collapse
|
15
|
Park A, Kim KE, Park I, Lee SH, Park KY, Jung M, Li X, Sleiman MB, Lee SJ, Kim DS, Kim J, Lim DS, Woo EJ, Lee EW, Han BS, Oh KJ, Lee SC, Auwerx J, Mun JY, Rhee HW, Kim WK, Bae KH, Suh JM. Mitochondrial matrix protein LETMD1 maintains thermogenic capacity of brown adipose tissue in male mice. Nat Commun 2023; 14:3746. [PMID: 37353518 PMCID: PMC10290150 DOI: 10.1038/s41467-023-39106-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 05/31/2023] [Indexed: 06/25/2023] Open
Abstract
Brown adipose tissue (BAT) has abundant mitochondria with the unique capability of generating heat via uncoupled respiration. Mitochondrial uncoupling protein 1 (UCP1) is activated in BAT during cold stress and dissipates mitochondrial proton motive force generated by the electron transport chain to generate heat. However, other mitochondrial factors required for brown adipocyte respiration and thermogenesis under cold stress are largely unknown. Here, we show LETM1 domain-containing protein 1 (LETMD1) is a BAT-enriched and cold-induced protein required for cold-stimulated respiration and thermogenesis of BAT. Proximity labeling studies reveal that LETMD1 is a mitochondrial matrix protein. Letmd1 knockout male mice display aberrant BAT mitochondria and fail to carry out adaptive thermogenesis under cold stress. Letmd1 knockout BAT is deficient in oxidative phosphorylation (OXPHOS) complex proteins and has impaired mitochondrial respiration. In addition, BAT-specific Letmd1 deficient mice exhibit phenotypes identical to those observed in Letmd1 knockout mice. Collectively, we demonstrate that the BAT-enriched mitochondrial matrix protein LETMD1 plays a tissue-autonomous role that is essential for BAT mitochondrial function and thermogenesis.
Collapse
Affiliation(s)
- Anna Park
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Kwang-Eun Kim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Isaac Park
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang Heon Lee
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Kun-Young Park
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Minkyo Jung
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, 41068, Republic of Korea
| | - Xiaoxu Li
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Maroun Bou Sleiman
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Su Jeong Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Dae-Soo Kim
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
- Digital Biotech Innovation Center, KRIBB, Daejeon, 34141, Republic of Korea
| | - Jaehoon Kim
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Dae-Sik Lim
- National Creative Research Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Eui-Jeon Woo
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
- Disease Target Structure Research Center, KRIBB, Daejeon, 34141, Republic of Korea
| | - Eun Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Baek Soo Han
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
- Biodefense Research Center, KRIBB, Daejeon, 34141, Republic of Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Ji Young Mun
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, 41068, Republic of Korea
| | - Hyun-Woo Rhee
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea.
- School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, CA, 92697, USA.
| | - Jae Myoung Suh
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
16
|
Arianti R, Ágnes Vinnai B, Győry F, Guba A, Csősz É, Kristóf E, Fésüs L. Availability of abundant thiamine determines efficiency of thermogenic activation in human neck area derived adipocytes. J Nutr Biochem 2023:109385. [PMID: 37230255 DOI: 10.1016/j.jnutbio.2023.109385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/23/2022] [Accepted: 05/20/2023] [Indexed: 05/27/2023]
Abstract
Brown/beige adipocytes express uncoupling protein-1 (UCP1) that enables them to dissipate energy as heat. Systematic activation of this process can alleviate obesity. Human brown adipose tissues are interspersed in distinct anatomical regions including deep neck. We found that UCP1 enriched adipocytes differentiated from precursors of this depot highly expressed ThTr2 transporter of thiamine and consumed thiamine during thermogenic activation of these adipocytes by cAMP which mimics adrenergic stimulation. Inhibition of ThTr2 led to lower thiamine consumption with decreased proton leak respiration reflecting reduced uncoupling. In the absence of thiamine, cAMP-induced uncoupling was diminished but restored by thiamine addition reaching the highest levels at thiamine concentrations larger than present in human blood plasma. Thiamine is converted to thiamine pyrophosphate (TPP) in cells; the addition of TPP to permeabilized adipocytes increased uncoupling fueled by TPP-dependent pyruvate dehydrogenase. ThTr2 inhibition also hampered cAMP-dependent induction of UCP1, PGC1a, and other browning marker genes, and thermogenic induction of these genes was potentiated by thiamine in a concentration dependent manner. Our study reveals the importance of amply supplied thiamine during thermogenic activation in human adipocytes which provides TPP for TPP-dependent enzymes not fully saturated with this cofactor and by potentiating the induction of thermogenic genes.
Collapse
Affiliation(s)
- Rini Arianti
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, H-4032, Debrecen, Hungary
| | - Boglárka Ágnes Vinnai
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, H-4032, Debrecen, Hungary
| | - Ferenc Győry
- Department of Surgery, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary
| | - Andrea Guba
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, H-4032, Debrecen, Hungary
| | - Éva Csősz
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary
| | - Endre Kristóf
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary.
| | - László Fésüs
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary.
| |
Collapse
|
17
|
Onikanni SA, Yang CY, Noriega L, Wang CH. U0126 Compound Triggers Thermogenic Differentiation in Preadipocytes via ERK-AMPK Signaling Axis. Int J Mol Sci 2023; 24:ijms24097987. [PMID: 37175694 PMCID: PMC10178890 DOI: 10.3390/ijms24097987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
In recent years, thermogenic differentiation and activation in brown and white adipose tissues have been regarded as one of the major innovative and promising strategies for the treatment and amelioration of obesity. However, the pharmacological approach towards this process has had limited and insufficient commitments, which presents a greater challenge for obesity treatment. This research evaluates the effects of U0126 compound on the activation of thermogenic differentiation during adipogenesis. The results show that U0126 pretreatment primes both white and brown preadipocytes to upregulate thermogenic and mitochondrial genes as well as enhance functions during the differentiation process. We establish that U0126-mediated thermogenic differentiation induction occurs partially via AMPK activation signaling. The findings of this research suggest U0126 as a promising alternative ligand in pursuit of a pharmacological option to increase thermogenic adipocyte formation and improve energy expenditure. Thus it could pave the way for the discovery of therapeutic drugs for the treatment of obesity and its related complications.
Collapse
Affiliation(s)
- Sunday Amos Onikanni
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
- Department of Chemical Sciences, Biochemistry Unit, Afe Babalola University, Ado-Ekiti 360101, Ekiti State, Nigeria
| | - Cheng-Ying Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Lloyd Noriega
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Chih-Hao Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
- Graduate Institute of Cell Biology, College of Life Sciences, China Medical University, Taichung 406040, Taiwan
| |
Collapse
|
18
|
Nicholls DG, Brand MD. A critical assessment of the role of creatine in brown adipose tissue thermogenesis. Nat Metab 2023; 5:21-28. [PMID: 36624158 DOI: 10.1038/s42255-022-00718-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/30/2022] [Indexed: 01/11/2023]
Abstract
Brown adipose tissue is specialized for non-shivering thermogenesis, combining lipolysis with an extremely active mitochondrial electron transport chain and a unique regulated uncoupling protein, UCP1, allowing unrestricted respiration. Current excitement focuses on the presence of brown adipose tissue in humans and the possibility that it may contribute to diet-induced thermogenesis, countering obesity and obesity-related disease as well as protecting cardio-metabolic health. In common with other tissues displaying a high, variable respiration, the tissue possesses a creatine pool and mitochondrial and cytosolic creatine kinase isoforms. Genetic and pharmacological manipulation of these components have pleiotropic effects that appear to influence diet- and cold-induced metabolism in vivo and modeled in vitro. These findings have been used to advance the concept of a UCP1-independent diet-induced thermogenic mechanism based on a dissipative hydrolysis of phosphocreatine in beige and brown adipose tissue. Here we review the in vivo and in vitro experimental basis for this hypothesis, and explore alternative explanations. We conclude that there is currently no convincing evidence for a significant futile creatine cycle in these tissues.
Collapse
|
19
|
Gu S, Gong Z, Liu S, Lu G, Ling Y, Wei Y, Li T, Gu R, Rong Y, Li J, Li H. Global Single-Cell Sequencing Landscape of Adipose Tissue of Different Anatomical Site Origin in Humans. Stem Cells Int 2023; 2023:8282961. [PMID: 37197688 PMCID: PMC10185425 DOI: 10.1155/2023/8282961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/28/2022] [Accepted: 03/20/2023] [Indexed: 05/19/2023] Open
Abstract
Chronic refractory wounds (CRW) are one of the most serious clinical challenges for surgeons to address. Stromal vascular fraction gels (SVFG), including human adipose stem cells (hASCs), have excellent vascular regenerative and tissue repair properties. Here, we combined single-cell RNA sequencing (scRNA-seq) of leg subcutaneous adipose tissue samples with scRNA-seq data from abdominal subcutaneous adipose tissue, leg subcutaneous adipose tissue, and visceral adipose tissue samples from public databases. The results showed specific differences in cellular levels in adipose tissue from different anatomical site sources. We identified cells including CD4+ T cells, hASCs, adipocyte (APC), epithelial (Ep) cells, and preadipocyte. In particular, the dynamics between groups of hASCs, epithelial cells, APCs, and precursor cells in adipose tissue of different anatomical site origins were more significant. Furthermore, our analysis reveals alterations at the cellular level and molecular level, as well as the biological signaling pathways involved in these subpopulations of cells with specific alterations. In particular, certain subpopulations of hASCs have higher cell stemness, which may be related to lipogenic differentiation capacity and may be beneficial in promoting CRW treatment and healing. In general, our study captures a human single-cell transcriptome profile across adipose depots, the cell type identification and analysis of which may help dissect the function and role of cells with specific alterations present in adipose tissue and may provide new ideas and approaches for the treatment of CRW in the clinical setting.
Collapse
Affiliation(s)
- Shixing Gu
- Department of Plastic and Aesthetic Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533022 Guangxi, China
| | - Zhenyu Gong
- Department of Burn, Plastic and Aesthetic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001 Guangxi, China
| | - Shuncai Liu
- Department of Burn, Plastic and Aesthetic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001 Guangxi, China
| | - Guohao Lu
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 53002, China
| | - Yu Ling
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 53002, China
| | - Yanlin Wei
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 53002, China
| | - Ting Li
- Department of Basic Science, YuanDong International Academy of Life Sciences, Hong Kong 999077, China
| | - Ronghe Gu
- Department of Orthopedics, The Fifth Affiliated Hospital of Guangxi Medical University & The First People's Hospital of Nanning, Nanning, 53002 Guangxi, China
| | - Yongxian Rong
- Department of Burn, Plastic and Aesthetic Surgery, The Guiping People's Hospital, Guigping, 537200, China
| | - Junjun Li
- Department of Pediatrics, The People's Hospital of Guangxi Zhuang Autonomous Region & Institute of Hospital Management and Medical Prevention Collaborative Innovation, Guangxi Academy of Medical Sciences, Nanning, 530021 Guangxi, China
| | - Hongmian Li
- Department of Plastic and Reconstructive Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region & Research Center of Medical Sciences, Guangxi Academy of Medical Sciences, Nanning, 530021 Guangxi, China
| |
Collapse
|
20
|
Guardiola-Márquez CE, Santos-Ramírez MT, Segura-Jiménez ME, Figueroa-Montes ML, Jacobo-Velázquez DA. Fighting Obesity-Related Micronutrient Deficiencies through Biofortification of Agri-Food Crops with Sustainable Fertilization Practices. PLANTS (BASEL, SWITZERLAND) 2022; 11:3477. [PMID: 36559589 PMCID: PMC9784404 DOI: 10.3390/plants11243477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 06/17/2023]
Abstract
Obesity is a critical medical condition worldwide that is increasingly involved with nutritional derangements associated with micronutrient deficiencies, including iron, zinc, calcium, magnesium, selenium, and vitamins A, C, D, and E. Nutritional deficiencies in obesity are mainly caused by poor-quality diets, higher nutrient requirements, alterations in micronutrient metabolism, and invasive obesity treatments. The current conventional agricultural system is designed for intensive food production, focusing on food quantity rather than food quality, consuming excessive agricultural inputs, and producing nutrient-deficient foods, thus generating severe health and environmental problems; agricultural food products may worsen obesity-related malnutrition. Therefore, modern agriculture is adopting new biofortification technologies to combat micronutrient deficiencies and improve agricultural productivity and sustainability. Biofertilization and nanofertilization practices are increasingly used due to their efficiency, safety, and reduced environmental impact. Biofertilizers are preparations of PGP-microorganisms that promote plant growth by influencing plant metabolism and improving the nutrient uptake, and nanofertilizers consist of synthesized nanoparticles with unique physicochemical properties that are capable of increasing plant nutrition and enriching agricultural products. This review presents the current micronutrient deficiencies associated with obesity, the modern unsustainable agri-food system contributing to obesity progression, and the development of bio- and nanofertilizers capable of biofortifying agri-food crops with micronutrients commonly deficient in patients with obesity.
Collapse
Affiliation(s)
| | - María Teresa Santos-Ramírez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. General Ramon Corona 2514, Zapopan 45138, Jalisco, Mexico
| | - M. Eugenia Segura-Jiménez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. General Ramon Corona 2514, Zapopan 45138, Jalisco, Mexico
| | - Melina Lizeth Figueroa-Montes
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. General Ramon Corona 2514, Zapopan 45138, Jalisco, Mexico
| | - Daniel A. Jacobo-Velázquez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. General Ramon Corona 2514, Zapopan 45138, Jalisco, Mexico
- Tecnologico de Monterrey, The Institute for Obesity Research, Ave. General Ramon Corona 2514, Zapopan 45201, Jalisco, Mexico
| |
Collapse
|
21
|
Guardiola-Márquez CE, Jacobo-Velázquez DA. Potential of enhancing anti-obesogenic agriceuticals by applying sustainable fertilizers during plant cultivation. FRONTIERS IN SUSTAINABLE FOOD SYSTEMS 2022. [DOI: 10.3389/fsufs.2022.1034521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Overweight and obesity are two of the world's biggest health problems. They are associated with excessive fat accumulation resulting from an imbalance between energy consumed and energy expended. Conventional therapies for obesity commonly include synthetic drugs and surgical procedures that can lead to serious side effects. Therefore, developing effective, safe, and readily available new treatments to prevent and treat obesity is highly relevant. Many plant extracts have shown anti-obesogenic potential. These plant extracts are composed of different agriceuticals such as fibers, phenolic acids, flavonoids, anthocyanins, alkaloids, lignans, and proteins that can manage obesity by suppressing appetite, inhibiting digestive enzymes, reducing adipogenesis and lipogenesis, promoting lipolysis and thermogenesis, modulating gut microbiota and suppressing obesity-induced inflammation. These anti-obesogenic agriceuticals can be enhanced in plants during their cultivation by applying sustainable fertilization strategies, improving their capacity to fight the obesity pandemic. Biofertilization and nanofertilization are considered efficient, eco-friendly, and cost-effective strategies to enhance plant growth and development and increase the content of nutrients and bioactive compounds, representing an alternative to overproducing the anti-obesogenic agriceuticals of interest. However, further research is required to study the impact of anti-obesogenic plant species grown using these agricultural practices. This review presents the current scenario of overweight and obesity; recent research work describing different plant species with significant effects against obesity; and several reports exhibiting the potential of the biofertilization and nanofertilization practices to enhance the concentrations of bioactive molecules of anti-obesogenic plant species.
Collapse
|
22
|
Du H, Shi L, Wang Q, Yan T, Wang Y, Zhang X, Yang C, Zhao Y, Yang X. Fu Brick Tea Polysaccharides Prevent Obesity via Gut Microbiota-Controlled Promotion of Adipocyte Browning and Thermogenesis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:13893-13903. [PMID: 36264038 DOI: 10.1021/acs.jafc.2c04888] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The antiobesity efficacy and underlying mechanisms of polysaccharides extracted from Fu brick tea (FBTP) were investigated. An 8-week administration of FBTP dose-dependently inhibited increases in body weight and weights of the epididymal-, retroperitoneal- and inguinal-white adipose tissues and stimulated beige-fat development and brown adipose tissue-derived nonshivering thermogenesis in high-fat diet-induced obese mice. FBTP protected against obesity-associated abnormality in serum adiponectin and leptin, indicating its positive regulation of energy metabolism. FBTP reversed gut dysbiosis by enriching beneficial bacteria, for example, Lactobacillus, Parabacteroides, Akkermansia, Bifidobacterium, and Roseburia. Results from the fecal microbiota transplantation further confirmed that FBTP-induced microbial shifts contributed to adipose browning and thermogenesis, thereby alleviating host adiposity, glucose homeostasis, dyslipidemia, and its related hepatic steatosis. Our study demonstrates the great potential of FBTP with prebiotic-like activities in preventing diet-induced obesity and its related metabolic complications via gut microbiota-derived enhancement of fat burning and energy expenditures.
Collapse
Affiliation(s)
- Haiping Du
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Lin Shi
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Qi Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Tao Yan
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yu Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xiangnan Zhang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Chengcheng Yang
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yan Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| |
Collapse
|
23
|
Al-Obaidi ZAF, Erdogan CS, Sümer E, Özgün HB, Gemici B, Sandal S, Yilmaz B. Investigation of obesogenic effects of hexachlorobenzene, DDT and DDE in male rats. Gen Comp Endocrinol 2022; 327:114098. [PMID: 35878704 DOI: 10.1016/j.ygcen.2022.114098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/04/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022]
Abstract
Obesity has become a very important public health problem and is increasing globally. Genetics, individual and environmental factors play roles in the etiology of this complex disorder. Recently, several environmental pollutants have been suggested to have obesogenic activities. Peroxisome proliferator activating receptor gamma (PPARγ), uncoupling protein-1 (UCP1) and their expression in white adipose tissue (WAT) and brown adipose tissue (BAT) play key roles in adipogenesis. UCP3 and irisin were reported to play roles in non-shivering thermogenesis. Our primary aim was to investigate obesogenic effects of hexachlorobenzene (HCB), dichlorodiphenyltrichloroethane (DDT) and dichlorodiphenyldichloroethylene (DDE) in rats. In addition, thermoregulatory effects of HCB, DDT and DDE were also investigated by analyzing the levels of Ucp3 and irisin. Thirty-two adult male Sprague-Dawley rats were randomly divided into four groups as control, HCB, DDT and DDE. Animals were administered with organochlorine pesticides (OCPs; 5 mg/kg bw) by oral gavage every other day for five weeks. At the end of the experimental period, the animals were sacrificed, BAT and WAT samples were collected to analyze Pparγ, Ucp1 and Ucp3 levels. Moreover, skeletal muscle samples were collected to examine Ucp3 and irisin levels. Serum glucose, cholesterol and triglyceride levels were also determined. Body weight and core temperature of the animals were not significantly affected by any of the OCP administration. Serum glucose, cholesterol and triglyceride levels were similar among the experimental groups. Pparγ expression was significantly elevated by HCB administration only in WAT (p < 0.05). On the other hand, both Pparγ and Ucp1 expressions were diminished in WAT and BAT (p < 0.01) by DDT treatment, while in WAT, DDE significantly decreased Pparγ expression without altering its expression in BAT (p < 0.001). Ucp3 and irisin levels in skeletal muscle were not altered. Our findings show that both DDT and DDE reduce the browning of WAT by suppressing white adipocytes and thus may have obesogenic activity in male rats without altering thermoregulation. In addition, HCB, DDT and DDE-induced alterations in expression of Pparγ and Ucp1 in WAT implicates differential regulation of adipogenic processes.
Collapse
Affiliation(s)
| | | | - Engin Sümer
- Yeditepe University, Faculty of Medicine, Experimental Research Center, Istanbul, Turkey
| | - Hüseyin Bugra Özgün
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Turkey
| | - Burcu Gemici
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Turkey
| | - Süleyman Sandal
- İnönü University, Faculty of Medicine, Department of Physiology, Malatya, Turkey
| | - Bayram Yilmaz
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Turkey.
| |
Collapse
|
24
|
Yin X, Chen Y, Ruze R, Xu R, Song J, Wang C, Xu Q. The evolving view of thermogenic fat and its implications in cancer and metabolic diseases. Signal Transduct Target Ther 2022; 7:324. [PMID: 36114195 PMCID: PMC9481605 DOI: 10.1038/s41392-022-01178-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 02/07/2023] Open
Abstract
AbstractThe incidence of metabolism-related diseases like obesity and type 2 diabetes mellitus has reached pandemic levels worldwide and increased gradually. Most of them are listed on the table of high-risk factors for malignancy, and metabolic disorders systematically or locally contribute to cancer progression and poor prognosis of patients. Importantly, adipose tissue is fundamental to the occurrence and development of these metabolic disorders. White adipose tissue stores excessive energy, while thermogenic fat including brown and beige adipose tissue dissipates energy to generate heat. In addition to thermogenesis, beige and brown adipocytes also function as dynamic secretory cells and a metabolic sink of nutrients, like glucose, fatty acids, and amino acids. Accordingly, strategies that activate and expand thermogenic adipose tissue offer therapeutic promise to combat overweight, diabetes, and other metabolic disorders through increasing energy expenditure and enhancing glucose tolerance. With a better understanding of its origins and biological functions and the advances in imaging techniques detecting thermogenesis, the roles of thermogenic adipose tissue in tumors have been revealed gradually. On the one hand, enhanced browning of subcutaneous fatty tissue results in weight loss and cancer-associated cachexia. On the other hand, locally activated thermogenic adipocytes in the tumor microenvironment accelerate cancer progression by offering fuel sources and is likely to develop resistance to chemotherapy. Here, we enumerate current knowledge about the significant advances made in the origin and physiological functions of thermogenic fat. In addition, we discuss the multiple roles of thermogenic adipocytes in different tumors. Ultimately, we summarize imaging technologies for identifying thermogenic adipose tissue and pharmacologic agents via modulating thermogenesis in preclinical experiments and clinical trials.
Collapse
|
25
|
Abstract
In this issue of Journal of Experimental Medicine, Yin et al. (2022. J. Exp. Med.https://doi.org/10.1084/jem.20212491) discover that loss of FNIP1 is associated with browning of white adipose tissue, which they propose is driven by decreased calcium uptake into the ER.
Collapse
Affiliation(s)
- Jakub Bunk
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Lawrence Kazak
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
26
|
Jiang N, Yang M, Han Y, Zhao H, Sun L. PRDM16 Regulating Adipocyte Transformation and Thermogenesis: A Promising Therapeutic Target for Obesity and Diabetes. Front Pharmacol 2022; 13:870250. [PMID: 35462933 PMCID: PMC9024053 DOI: 10.3389/fphar.2022.870250] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Given that obesity and diabetes have been major public health concerns and that disease morbidities have been rising continuously, effective treatment for these diseases is urgently needed. Because adipose tissue metabolism is involved in the progression of obesity and diabetes, it might be efficient to target adipocyte metabolic pathways. Positive regulatory domain zinc finger region protein 16 (PRDM16), a transcription factor that is highly expressed in adipocytes, plays a key role in adipose tissue metabolism, such as the browning and thermogenesis of adipocytes, the beigeing of adipocytes, the adipogenic differentiation of myoblasts, and the conversion of visceral adipocytes to subcutaneous adipocytes. Furthermore, clinical and basic studies have shown that the expression of PRDM16 is associated with obesity and diabetes and that PRDM16 signaling participates in the treatment of the two diseases. For example, metformin promotes thermogenesis and alleviates obesity by activating the AMPK/αKG/PRDM16 signaling pathway; rosiglitazone alleviates obesity under the synergistic effect of PRDM16; resveratrol plays an antiobesity role by inducing the expression of PRDM16; liraglupeptide improves insulin resistance by inducing the expression of PRDM16; and mulberry leaves play an anti-inflammatory and antidiabetes role by activating the expression of brown fat cell marker genes (including PRDM16). In this review, we summarize the evidence of PRDM16 involvement in the progression of obesity and diabetes and that PRDM16 may be a promising therapy for obesity and diabetes.
Collapse
|
27
|
Brock CK, Hebert KL, Artiles M, Wright MK, Cheng T, Windsor GO, Nguyen K, Alzoubi MS, Collins-Burow BM, Martin EC, Lau FH, Bunnell BA, Burow ME. A Role for Adipocytes and Adipose Stem Cells in the Breast Tumor Microenvironment and Regenerative Medicine. Front Physiol 2021; 12:751239. [PMID: 34912237 PMCID: PMC8667576 DOI: 10.3389/fphys.2021.751239] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022] Open
Abstract
Obesity rates are climbing, representing a confounding and contributing factor to many disease states, including cancer. With respect to breast cancer, obesity plays a prominent role in the etiology of this disease, with certain subtypes such as triple-negative breast cancer having a strong correlation between obesity and poor outcomes. Therefore, it is critical to examine the obesity-related alterations to the normal stroma and the tumor microenvironment (TME). Adipocytes and adipose stem cells (ASCs) are major components of breast tissue stroma that have essential functions in both physiological and pathological states, including energy storage and metabolic homeostasis, physical support of breast epithelial cells, and directing inflammatory and wound healing responses through secreted factors. However, these processes can become dysregulated in both metabolic disorders, such as obesity and also in the context of breast cancer. Given the well-established obesity-neoplasia axis, it is critical to understand how interactions between different cell types in the tumor microenvironment, including adipocytes and ASCs, govern carcinogenesis, tumorigenesis, and ultimately metastasis. ASCs and adipocytes have multifactorial roles in cancer progression; however, due to the plastic nature of these cells, they also have a role in regenerative medicine, making them promising tools for tissue engineering. At the physiological level, the interactions between obesity and breast cancer have been examined; here, we will delineate the mechanisms that regulate ASCs and adipocytes in these different contexts through interactions between cancer cells, immune cells, and other cell types present in the tumor microenvironment. We will define the current state of understanding of how adipocytes and ASCs contribute to tumor progression through their role in the tumor microenvironment and how this is altered in the context of obesity. We will also introduce recent developments in utilizing adipocytes and ASCs in novel approaches to breast reconstruction and regenerative medicine.
Collapse
Affiliation(s)
- Courtney K Brock
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Katherine L Hebert
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Maria Artiles
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Maryl K Wright
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Thomas Cheng
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Gabrielle O Windsor
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Khoa Nguyen
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Madlin S Alzoubi
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Bridgette M Collins-Burow
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Elizabeth C Martin
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Frank H Lau
- Section of Plastic & Reconstructive Surgery, Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Bruce A Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Matthew E Burow
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
28
|
Mishra BK, Madhu SV, Aslam M, Agarwal V, Banerjee BD. Adipose tissue expression of UCP1 and PRDM16 genes and their association with postprandial triglyceride metabolism and glucose intolerance. Diabetes Res Clin Pract 2021; 182:109115. [PMID: 34718051 DOI: 10.1016/j.diabres.2021.109115] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 08/28/2021] [Accepted: 10/15/2021] [Indexed: 12/01/2022]
Abstract
AIMS UCP1 and PRDM16 genes, primarily involved in browning of adipose tissue that can affect lipid metabolism are also associated with diabetes risk. Therefore, we planned to study the adipose tissue expression of UCP1 and PRDM 16 genes in subjects with glucose intolerance to find out its association with postprandial triglyceride (PPTg) measures and T2DM. METHODS A total of 30 subjects were recruited in three groups i.e., NGT, prediabetes and T2DM (NDDM + known T2DM) who were matched for age, sex and BMI. An 8-hour standardized fat challenge test was performed to study lipemic responses. UCP1 and PRDM16 genes quantification in adipose tissue was performed by real-time PCR followed by SDS PAGE. RESULTS UCP1 gene expression in SAT was significantly lower in T2DM and prediabetes as compared to NGT group while PRDM16 gene expression was significantly lower in T2DM group as compared to NGT group. UCP1 gene expression correlated with PPTg measures as well as with glycaemic measures while PRDM16 gene expression correlated with glycaemic measures only. CONCLUSION This study found downregulation of PRDM16 and UCP1 gene expression in SAT in subjects with glucose intolerance. The association of UCP1 gene expression with PPTg dysmetabolism may contribute to greater predisposition to T2DM.
Collapse
Affiliation(s)
- B K Mishra
- Department of Endocrinology, University College of Medical Sciences & GTB Hospital, University of Delhi, India
| | - S V Madhu
- Department of Endocrinology, University College of Medical Sciences & GTB Hospital, University of Delhi, India.
| | - M Aslam
- Department of Endocrinology, University College of Medical Sciences & GTB Hospital, University of Delhi, India
| | - V Agarwal
- Department of Surgery, University College of Medical Sciences & GTB Hospital, University of Delhi, India
| | - B D Banerjee
- Department of Biochemistry, University College of Medical Sciences & GTB Hospital, University of Delhi, India
| |
Collapse
|
29
|
BMP7 Increases UCP1-Dependent and Independent Thermogenesis with a Unique Gene Expression Program in Human Neck Area Derived Adipocytes. Pharmaceuticals (Basel) 2021; 14:ph14111078. [PMID: 34832860 PMCID: PMC8625022 DOI: 10.3390/ph14111078] [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] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/26/2022] Open
Abstract
White adipocytes contribute to energy storage, accumulating lipid droplets, whereas brown and beige adipocytes mainly function in dissipating energy as heat primarily via the action of uncoupling protein 1 (UCP1). Bone morphogenic protein 7 (BMP7) was shown to drive brown adipocyte differentiation in murine interscapular adipose tissue. Here, we performed global RNA-sequencing and functional assays on adipocytes obtained from subcutaneous (SC) and deep-neck (DN) depots of human neck and differentiated with or without BMP7. We found that BMP7 did not influence differentiation but upregulated browning markers, including UCP1 mRNA and protein in SC and DN derived adipocytes. BMP7 also enhanced mitochondrial DNA content, levels of oxidative phosphorylation complex subunits, along with PGC1α and p-CREB upregulation, and fragmentation of mitochondria. Furthermore, both UCP1-dependent proton leak and UCP1-independent, creatine-driven substrate cycle coupled thermogenesis were augmented upon BMP7 addition. The gene expression analysis also shed light on the possible role of genes unrelated to thermogenesis thus far, including ACAN, CRYAB, and ID1, which were among the highest upregulated ones by BMP7 treatment in both types of adipocytes. Together, our study shows that BMP7 strongly upregulates thermogenesis in human neck area derived adipocytes, along with genes, which might have a supporting role in energy expenditure.
Collapse
|
30
|
Choi C, Son Y, Kim J, Cho YK, Saha A, Kim M, Im H, Kim K, Han J, Lee JW, Seong JK, Lee YH. TM4SF5 Knockout Protects Mice From Diet-Induced Obesity Partly by Regulating Autophagy in Adipose Tissue. Diabetes 2021; 70:2000-2013. [PMID: 34187836 PMCID: PMC8576418 DOI: 10.2337/db21-0145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/22/2021] [Indexed: 11/13/2022]
Abstract
Transmembrane 4 L six family member 5 (TM4SF5) functions as a sensor for lysosomal arginine levels and activates the mammalian target of rapamycin complex 1 (mTORC1). While the mTORC1 signaling pathway plays a key role in adipose tissue metabolism, the regulatory function of TM4SF5 in adipocytes remains unclear. In this study we aimed to establish a TM4SF5 knockout (KO) mouse model and investigated the effects of TM4SF5 KO on mTORC1 signaling-mediated autophagy and mitochondrial metabolism in adipose tissue. TM4SF5 expression was higher in inguinal white adipose tissue (iWAT) than in brown adipose tissue and significantly upregulated by a high-fat diet (HFD). TM4SF5 KO reduced mTORC1 activation and enhanced autophagy and lipolysis in adipocytes. RNA sequencing analysis of TM4SF5 KO mouse iWAT showed that the expression of genes involved in peroxisome proliferator-activated receptor α signaling pathways and mitochondrial oxidative metabolism was upregulated. Consequently, TM4SF5 KO reduced adiposity and increased energy expenditure and mitochondrial oxidative metabolism. TM4SF5 KO prevented HFD-induced glucose intolerance and inflammation in adipose tissue. Collectively, the results of our study demonstrate that TM4SF5 regulates autophagy and lipid catabolism in adipose tissue and suggest that TM4SF5 could be therapeutically targeted for the treatment of obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Cheoljun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yeonho Son
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jinyoung Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoon Keun Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Abhirup Saha
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Minsu Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyeonyeong Im
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Kyungmin Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Juhyeong Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jung Weon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 Plus Program for Advanced Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, and Korea Mouse Phenotyping Center, Seoul National University, Seoul, Republic of Korea
| | - Yun-Hee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
31
|
Chronic Intermittent Hypoxia Induces Early-Stage Metabolic Dysfunction Independently of Adipose Tissue Deregulation. Antioxidants (Basel) 2021; 10:antiox10081233. [PMID: 34439481 PMCID: PMC8388878 DOI: 10.3390/antiox10081233] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/09/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
Several studies demonstrated a link between obstructive sleep apnea (OSA) and the development of insulin resistance. However, the main event triggering insulin resistance in OSA remains to be clarified. Herein, we investigated the effect of mild and severe chronic intermittent hypoxia (CIH) on whole-body metabolic deregulation and visceral adipose tissue dysfunction. Moreover, we studied the contribution of obesity to CIH-induced dysmetabolic states. Experiments were performed in male Wistar rats submitted to a control and high-fat (HF) diet. Two CIH protocols were tested: A mild CIH paradigm (5/6 hypoxic (5% O2) cycles/h, 10.5 h/day) during 35 days and a severe CIH paradigm (30 hypoxic (5% O2) cycles, 8 h/day) during 15 days. Fasting glycemia, insulinemia, insulin sensitivity, weight, and fat mass were assessed. Adipose tissue hypoxia, inflammation, angiogenesis, oxidative stress, and metabolism were investigated. Mild and severe CIH increased insulin levels and induced whole-body insulin resistance in control animals, effects not associated with weight gain. In control animals, CIH did not modify adipocytes perimeter as well as adipose tissue hypoxia, angiogenesis, inflammation or oxidative stress. In HF animals, severe CIH attenuated the increase in adipocytes perimeter, adipose tissue hypoxia, angiogenesis, and dysmetabolism. In conclusion, adipose tissue dysfunction is not the main trigger for initial dysmetabolism in CIH. CIH in an early stage might have a protective role against the deleterious effects of HF diet on adipose tissue metabolism.
Collapse
|
32
|
Lagarde D, Jeanson Y, Portais JC, Galinier A, Ader I, Casteilla L, Carrière A. Lactate Fluxes and Plasticity of Adipose Tissues: A Redox Perspective. Front Physiol 2021; 12:689747. [PMID: 34276410 PMCID: PMC8278056 DOI: 10.3389/fphys.2021.689747] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/09/2021] [Indexed: 12/18/2022] Open
Abstract
Lactate, a metabolite produced when the glycolytic flux exceeds mitochondrial oxidative capacities, is now viewed as a critical regulator of metabolism by acting as both a carbon and electron carrier and a signaling molecule between cells and tissues. In recent years, increasing evidence report its key role in white, beige, and brown adipose tissue biology, and highlights new mechanisms by which lactate participates in the maintenance of whole-body energy homeostasis. Lactate displays a wide range of biological effects in adipose cells not only through its binding to the membrane receptor but also through its transport and the subsequent effect on intracellular metabolism notably on redox balance. This study explores how lactate regulates adipocyte metabolism and plasticity by balancing intracellular redox state and by regulating specific signaling pathways. We also emphasized the contribution of adipose tissues to the regulation of systemic lactate metabolism, their roles in redox homeostasis, and related putative physiopathological repercussions associated with their decline in metabolic diseases and aging.
Collapse
Affiliation(s)
- Damien Lagarde
- Goodman Cancer Research Center, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada.,Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Yannick Jeanson
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Jean-Charles Portais
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France.,MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Anne Galinier
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France.,Institut Fédératif de Biologie, CHU Purpan, Toulouse, France
| | - Isabelle Ader
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Louis Casteilla
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Audrey Carrière
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
33
|
Cytochrome P450 2E1 (CYP2E1) positively regulates lipid catabolism and induces browning in 3T3-L1 white adipocytes. Life Sci 2021; 278:119648. [PMID: 34043994 DOI: 10.1016/j.lfs.2021.119648] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
AIMS Browning induction (beiging) of white adipocytes is an emerging prospective strategy to defeat obesity and its related metabolic disorders. Cytochrome P450 2E1 (CYP2E1), a membrane protein which belongs to the cytochrome P450 superfamily, reportedly functions in the xenobiotic metabolism in the body, especially ethanol metabolism. Although previous studies have reported the effect of CYP2E1 on obesity in animal models, the data remains controversial. In the current study, we investigate for the first time, the role of CYP2E1 in lipid metabolism in 3T3-L1 white adipocytes, with a focus on fat browning. METHODS 3T3-L1 white adipocytes and Cyp2e1 siRNA were applied to investigate the role of CYP2E1 in white adipocytes. After that, cells were seperately exposed to β3-AR agonist, β3-AR antagonist and p38 inhibitor to identify the pathway which CYP2E1 was involved in to regulate browning event in white adipocytes. KEY FINDINGS We found that CYP2E1 deficiency results in reduced adipogenesis and lipogenesis as well as brown adipocyte-like phenotype induction. A mechanistic study to identify the molecular signals for CYP2E1 regulation in the browning of white adipocytes revealed that CYP2E1 inhibition deters the β3-adrenergic receptor activation and its downstream targets. SIGNIFICANCE Our data unveilved a previously unknown mechanism in the regulation of browning by CYP2E1 in 3T3-L1 white adipocytes, suggesting that CYP2E1 is a promising molecular target for the treatment of obesity and its related diseases.
Collapse
|
34
|
Chen L, Liu R, He X, Pei S, Li D. Effects of brown seaweed polyphenols, a class of phlorotannins, on metabolic disorders via regulation of fat function. Food Funct 2021; 12:2378-2388. [PMID: 33645609 DOI: 10.1039/d0fo02886j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
It is well known that fat dysfunction is the main driver of development of metabolic disorders. Changes in diet and lifestyle are particularly important to reverse the current global rise in obesity-related metabolic disorders. Seaweed has been consumed for thousands of years, and it is rich in bioactive compounds, especially unique polyphenols. The aim of the present review is to summarize the effects of different seaweed polyphenols on fat function in metabolic disorders and the related mechanisms. Seaweed polyphenols activate white adipose tissue to "brown" or "beige" adipose tissue to enhance energy consumption. In addition, the amelioration of fat factor imbalance and inflammatory response is also considered as an important reason for the regulation of lipid function with seaweed polyphenols. The present review provides an important basis for using seaweed polyphenols as potential dietary supplements to prevent metabolic disorders.
Collapse
Affiliation(s)
- Lei Chen
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
| | | | | | | | | |
Collapse
|
35
|
Hammoud SH, AlZaim I, Al-Dhaheri Y, Eid AH, El-Yazbi AF. Perirenal Adipose Tissue Inflammation: Novel Insights Linking Metabolic Dysfunction to Renal Diseases. Front Endocrinol (Lausanne) 2021; 12:707126. [PMID: 34408726 PMCID: PMC8366229 DOI: 10.3389/fendo.2021.707126] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
A healthy adipose tissue (AT) is indispensable to human wellbeing. Among other roles, it contributes to energy homeostasis and provides insulation for internal organs. Adipocytes were previously thought to be a passive store of excess calories, however this view evolved to include an endocrine role. Adipose tissue was shown to synthesize and secrete adipokines that are pertinent to glucose and lipid homeostasis, as well as inflammation. Importantly, the obesity-induced adipose tissue expansion stimulates a plethora of signals capable of triggering an inflammatory response. These inflammatory manifestations of obese AT have been linked to insulin resistance, metabolic syndrome, and type 2 diabetes, and proposed to evoke obesity-induced comorbidities including cardiovascular diseases (CVDs). A growing body of evidence suggests that metabolic disorders, characterized by AT inflammation and accumulation around organs may eventually induce organ dysfunction through a direct local mechanism. Interestingly, perirenal adipose tissue (PRAT), surrounding the kidney, influences renal function and metabolism. In this regard, PRAT emerged as an independent risk factor for chronic kidney disease (CKD) and is even correlated with CVD. Here, we review the available evidence on the impact of PRAT alteration in different metabolic states on the renal and cardiovascular function. We present a broad overview of novel insights linking cardiovascular derangements and CKD with a focus on metabolic disorders affecting PRAT. We also argue that the confluence among these pathways may open several perspectives for future pharmacological therapies against CKD and CVD possibly by modulating PRAT immunometabolism.
Collapse
Affiliation(s)
- Safaa H. Hammoud
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Beirut Arab University, Beirut, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Departmment of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yusra Al-Dhaheri
- Department of Biology, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University (QU) Health, Qatar University, Doha, Qatar
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt
- *Correspondence: Ahmed F. El-Yazbi,
| |
Collapse
|
36
|
Poursharifi P, Attané C, Mugabo Y, Al-Mass A, Ghosh A, Schmitt C, Zhao S, Guida J, Lussier R, Erb H, Chenier I, Peyot ML, Joly E, Noll C, Carpentier AC, Madiraju SRM, Prentki M. Adipose ABHD6 regulates tolerance to cold and thermogenic programs. JCI Insight 2020; 5:140294. [PMID: 33201859 PMCID: PMC7819748 DOI: 10.1172/jci.insight.140294] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 11/11/2020] [Indexed: 12/31/2022] Open
Abstract
Enhanced energy expenditure in brown (BAT) and white adipose tissues (WAT) can be therapeutic against metabolic diseases. We examined the thermogenic role of adipose α/β-hydrolase domain 6 (ABHD6), which hydrolyzes monoacylglycerol (MAG), by employing adipose-specific ABHD6-KO mice. Control and KO mice showed similar phenotypes at room temperature and thermoneutral conditions. However, KO mice were resistant to hypothermia, which can be accounted for by the simultaneously increased lipolysis and lipogenesis of the thermogenic glycerolipid/free fatty acid (GL/FFA) cycle in visceral fat, despite unaltered uncoupling protein 1 expression. Upon cold stress, nuclear 2-MAG levels increased in visceral WAT of the KO mice. Evidence is provided that 2-MAG causes activation of PPARα in white adipocytes, leading to elevated expression and activity of GL/FFA cycle enzymes. In the ABHD6-ablated BAT, glucose and oxidative metabolism were elevated upon cold induction, without changes in GL/FFA cycle and lipid turnover. Moreover, response to in vivo β3-adrenergic stimulation was comparable between KO and control mice. Our data reveal a MAG/PPARα/GL/FFA cycling metabolic signaling network in visceral adipose tissue, which contributes to cold tolerance, and that adipose ABHD6 is a negative modulator of adaptive thermogenesis. Visceral adipose adipose α/β-hydrolase domain 6 regulates cold adaptation and acts as a brake for heat production via the regulation of thermogenic glycerolipid/free fatty acid cycling.
Collapse
Affiliation(s)
- Pegah Poursharifi
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Camille Attané
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Yves Mugabo
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Anfal Al-Mass
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Anindya Ghosh
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Clémence Schmitt
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Shangang Zhao
- Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Julian Guida
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Roxane Lussier
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Heidi Erb
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Isabelle Chenier
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Marie-Line Peyot
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Erik Joly
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Christophe Noll
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - S R Murthy Madiraju
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Marc Prentki
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| |
Collapse
|
37
|
Tanaka Y, Taguchi S, Maruyama K, Mori K, Miyazato M, Kangawa K, Murakami N, Nakahara K. Comparison of physiological functions between neuromedin U-related peptide and neuromedin S-related peptide in the rat central nervous system. Biochem Biophys Res Commun 2020; 534:653-658. [PMID: 33228964 DOI: 10.1016/j.bbrc.2020.11.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 11/08/2020] [Indexed: 02/02/2023]
Abstract
Two novel peptides, neuromedin U precursor-related peptide (NURP) and neuromedin S precursor-related peptide (NSRP), are produced from neuromedin U (NMU) and neuromedin S (NMS) precursors, respectively, as these precursors have multiple consensus sequences for proteolytic processing. Our group has shown previously that one of these two novel peptides, NURP, stimulates body temperature and locomotor activity, but not food intake. However, the physiological function of the other peptide, NSRP, has remained unclear. Therefore, the aim of this study was to characterize differences in the regions of the rat brain targeted by the NMU/NMS peptide family, including NURP and NSRP, and their physiological functions. First, we explored the regions of c-Fos expression after intracerebroventricular (i.c.v.) injection of NURP and NSRP and found that these were fewer than after i.c.v. injection of NMU and NMS in the hypothalamus, possibly because NURP and NSRP cannot activate NMU/NMS receptors. In the ventral subiculum, which is one region of the hippocampus, c-Fos expression was evident only after i.c.v. injection of NURP. We also examined the effects of NSRP on food intake, body temperature and locomotor activity. Like NURP, NSRP increased both body temperature and locomotor activity, but not food intake, indicating that NSRP is also a functional peptide. However, these effects of NSRP were distinctly weaker than those of NURP. These findings suggest differences in the affinity of NURP and/or NSRP for specific receptors, or in their respective biological activities.
Collapse
Affiliation(s)
- Yukie Tanaka
- Laboratory of Veterinary Physiology, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadainishi, Miyazaki, Miyazaki 889-2192, Japan
| | - Shimon Taguchi
- Laboratory of Veterinary Physiology, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadainishi, Miyazaki, Miyazaki 889-2192, Japan
| | - Keisuke Maruyama
- Laboratory of Veterinary Physiology, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadainishi, Miyazaki, Miyazaki 889-2192, Japan.
| | - Kenji Mori
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka, 564-8565, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka, 564-8565, Japan
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka, 564-8565, Japan
| | - Noboru Murakami
- Laboratory of Veterinary Physiology, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadainishi, Miyazaki, Miyazaki 889-2192, Japan
| | - Keiko Nakahara
- Laboratory of Veterinary Physiology, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadainishi, Miyazaki, Miyazaki 889-2192, Japan
| |
Collapse
|
38
|
Lizcano F, Arroyave F. Control of Adipose Cell Browning and Its Therapeutic Potential. Metabolites 2020; 10:metabo10110471. [PMID: 33227979 PMCID: PMC7699191 DOI: 10.3390/metabo10110471] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/20/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
Adipose tissue is the largest endocrine organ in humans and has an important influence on many physiological processes throughout life. An increasing number of studies have described the different phenotypic characteristics of fat cells in adults. Perhaps one of the most important properties of fat cells is their ability to adapt to different environmental and nutritional conditions. Hypothalamic neural circuits receive peripheral signals from temperature, physical activity or nutrients and stimulate the metabolism of white fat cells. During this process, changes in lipid inclusion occur, and the number of mitochondria increases, giving these cells functional properties similar to those of brown fat cells. Recently, beige fat cells have been studied for their potential role in the regulation of obesity and insulin resistance. In this context, it is important to understand the embryonic origin of beige adipocytes, the response of adipocyte to environmental changes or modifications within the body and their ability to transdifferentiate to elucidate the roles of these cells for their potential use in therapeutic strategies for obesity and metabolic diseases. In this review, we discuss the origins of the different fat cells and the possible therapeutic properties of beige fat cells.
Collapse
Affiliation(s)
- Fernando Lizcano
- Center of Biomedical Investigation, (CIBUS), Universidad de La Sabana, 250008 Chia, Colombia
- Correspondence:
| | - Felipe Arroyave
- Doctoral Program in Biociencias, Universidad de La Sabana, 250008 Chia, Colombia
| |
Collapse
|
39
|
Colson C, Batrow PL, Gautier N, Rochet N, Ailhaud G, Peiretti F, Amri EZ. The Rosmarinus Bioactive Compound Carnosic Acid Is a Novel PPAR Antagonist That Inhibits the Browning of White Adipocytes. Cells 2020; 9:cells9112433. [PMID: 33171828 PMCID: PMC7695189 DOI: 10.3390/cells9112433] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022] Open
Abstract
Thermogenic brown and brite adipocytes convert chemical energy from nutrients into heat. Therapeutics that regulate brown adipocyte recruitment and activity represent interesting strategies to control fat mass such as in obesity or cachexia. The peroxisome proliferator-activated receptor (PPAR) family plays key roles in the maintenance of adipose tissue and in the regulation of thermogenic activity. Activation of these receptors induce browning of white adipocyte. The purpose of this work was to characterize the role of carnosic acid (CA), a compound used in traditional medicine, in the control of brown/brite adipocyte formation and function. We used human multipotent adipose-derived stem (hMADS) cells differentiated into white or brite adipocytes. The expression of key marker genes was determined using RT-qPCR and western blotting. We show here that CA inhibits the browning of white adipocytes and favors decreased gene expression of thermogenic markers. CA treatment does not affect β-adrenergic response. Importantly, the effects of CA are fully reversible. We used transactivation assays to show that CA has a PPARα/γ antagonistic action. Our data pinpoint CA as a drug able to control PPAR activity through an antagonistic effect. These observations shed some light on the development of natural PPAR antagonists and their potential effects on thermogenic response.
Collapse
Affiliation(s)
- Cécilia Colson
- Université Côte d’Azur, CNRS, Inserm, iBV, 06103 Nice, France; (C.C.); (P.-L.B.); (N.G.); (N.R.); (G.A.)
| | - Pierre-Louis Batrow
- Université Côte d’Azur, CNRS, Inserm, iBV, 06103 Nice, France; (C.C.); (P.-L.B.); (N.G.); (N.R.); (G.A.)
| | - Nadine Gautier
- Université Côte d’Azur, CNRS, Inserm, iBV, 06103 Nice, France; (C.C.); (P.-L.B.); (N.G.); (N.R.); (G.A.)
| | - Nathalie Rochet
- Université Côte d’Azur, CNRS, Inserm, iBV, 06103 Nice, France; (C.C.); (P.-L.B.); (N.G.); (N.R.); (G.A.)
| | - Gérard Ailhaud
- Université Côte d’Azur, CNRS, Inserm, iBV, 06103 Nice, France; (C.C.); (P.-L.B.); (N.G.); (N.R.); (G.A.)
| | - Franck Peiretti
- Aix Marseille Université, INSERM, INRAE, C2VN, 13007 Marseille, France;
| | - Ez-Zoubir Amri
- Université Côte d’Azur, CNRS, Inserm, iBV, 06103 Nice, France; (C.C.); (P.-L.B.); (N.G.); (N.R.); (G.A.)
- Correspondence: ; Tel.: +33-493-37-70-82; Fax: +33-493-81-70-58
| |
Collapse
|
40
|
Bartke A, Brannan S, Hascup E, Hascup K, Darcy J. Energy Metabolism and Aging. World J Mens Health 2020; 39:222-232. [PMID: 33151044 PMCID: PMC7994661 DOI: 10.5534/wjmh.200112] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/04/2020] [Accepted: 08/28/2020] [Indexed: 12/21/2022] Open
Abstract
Aging is strongly related to energy metabolism, but the underlying processes and mechanisms are complex and incompletely understood. Restricting energy intake and reducing metabolic rate can slow the rate of aging and extend longevity, implying a reciprocal relationship between energy metabolism and life expectancy. However, increased energy expenditure has also been associated with improved health and longer life. In both experimental animals and humans, reduced body temperature has been related to extended longevity. However, recent findings on the function of thermogenic (brown or beige) adipose tissue produced intense interest in increasing the amount of energy expended for thermogenesis to prevent and/or treat obesity, improve metabolic health, and extend life. Evidence available to-date indicates that increasing adipose tissue thermogenesis by pharmacologic, environmental, or genetic interventions can indeed produce significant metabolic benefits, which are associated with improved chances for healthy aging and long life.
Collapse
Affiliation(s)
- Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA.
| | - Savannah Brannan
- Department of Biology, University of Illinois Springfield, Springfield, IL, USA
| | - Erin Hascup
- Department of Neurology and Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin Hascup
- Department of Neurology and Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Justin Darcy
- Joslin Diabetes Center, Section on Integrative Physiology and Metabolism, Harvard Medical School One Joslin Place, Boston, MA, USA
| |
Collapse
|
41
|
Dugail I, Amri EZ, Vitale N. High prevalence for obesity in severe COVID-19: Possible links and perspectives towards patient stratification. Biochimie 2020; 179:257-265. [PMID: 32649962 PMCID: PMC7340594 DOI: 10.1016/j.biochi.2020.07.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022]
Abstract
It is becoming obvious that in addition to aging and various hearth pathologies, excess of body weight, especially obesity is a major risk factor for severity of COVID-19 infection. Intriguingly the receptor for SARS-CoV-2 is ACE2, a member of the angiotensin receptor family that has a relatively large tissue distribution. This observation likely explains the multitude of symptoms that have been described from human patients. The adipose tissue also expresses ACE2, suggesting that adipocytes are potentially infected by SARS-CoV-2. Here we discuss some of the potential contribution of the adipose tissue to the severity of the infection and propose some aspects of obese patients metabolic phenotyping to help stratification of individuals with high risk of severe disease.
Collapse
Affiliation(s)
- Isabelle Dugail
- UMRS 1269 INSERM/Sorbonne University, Nutriomics, Faculté de médecine Pitié-Salpêtrière, F-75013, Paris, France; Member of the scientific council of the Groupe d'Etude et de Recherche en Lipidomique (GERLI), France
| | - Ez-Zoubir Amri
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France; Member of the scientific council of the Groupe d'Etude et de Recherche en Lipidomique (GERLI), France
| | - Nicolas Vitale
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, F-67000, Strasbourg, France; Member of the scientific council of the Groupe d'Etude et de Recherche en Lipidomique (GERLI), France.
| |
Collapse
|
42
|
Coleman RA. The "discovery" of lipid droplets: A brief history of organelles hidden in plain sight. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158762. [PMID: 32622088 DOI: 10.1016/j.bbalip.2020.158762] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022]
Abstract
Mammalian lipid droplets (LDs), first described as early as the 1880s, were virtually ignored for more than 100 years. Between 1991 and the early 2000s, however, a series of discoveries and conceptual breakthroughs led to a resurgent interest in obesity as a disease, in the metabolism of intracellular triacylglycerol (TAG), and in the physical locations of LDs as cellular structures with their associated proteins. Insights included the recognition that obesity underlies major chronic diseases, that appetite is hormonally controlled, that hepatic steatosis is not a benign finding, and that diabetes might fundamentally be a disorder of lipid metabolism. In this brief review, I describe the metamorphosis of LDs from overlooked globs of stored fat to dynamic organelles that control insulin resistance, mitochondrial oxidation, and viral replication.
Collapse
Affiliation(s)
- Rosalind A Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America.
| |
Collapse
|