1
|
Thakur P, Baraskar K, Shrivastava VK, Medhi B. Cross-talk between adipose tissue and microbiota-gut-brain-axis in brain development and neurological disorder. Brain Res 2024; 1844:149176. [PMID: 39182900 DOI: 10.1016/j.brainres.2024.149176] [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: 04/12/2024] [Revised: 07/25/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
The gut microbiota is an important factor responsible for the physiological processes as well as pathogenesis of host. The communication between central nervous system (CNS) and microbiota occurs by different pathways i.e., chemical, neural, immune, and endocrine. Alteration in gut microbiota i.e., gut dysbiosis causes alteration in the bidirectional communication between CNS and gut microbiota and linked to the pathogenesis of neurological and neurodevelopmental disorder. Therefore, now-a-days microbiota-gut-brain-axis (MGBA) has emerged as therapeutic target for the treatment of metabolic disorder. But, experimental data available on MGBA from basic research has limited application in clinical study. In present study we first summarized molecular mechanism of microbiota interaction with brain physiology and pathogenesis via collecting data from different sources i.e., PubMed, Scopus, Web of Science. Furthermore, evidence shows that adipose tissue (AT) is active during metabolic activities and may also interact with MGBA. Hence, in present study we have focused on the relationship among MGBA, brown adipose tissue, and white adipose tissue. Along with this, we have also studied functional specificity of AT, and understanding heterogeneity among MGBA and different types of AT. Therefore, molecular interaction among them may provide therapeutic target for the treatment of neurological disorder.
Collapse
Affiliation(s)
- Pratibha Thakur
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India.
| | - Kirti Baraskar
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Vinoy K Shrivastava
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, Punjab 160012, India.
| |
Collapse
|
2
|
Rezq S, Huffman AM, Basnet J, Alsemeh AE, do Carmo JM, Yanes Cardozo LL, Romero DG. MicroRNA-21 modulates brown adipose tissue adipogenesis and thermogenesis in a mouse model of polycystic ovary syndrome. Biol Sex Differ 2024; 15:53. [PMID: 38987854 PMCID: PMC11238487 DOI: 10.1186/s13293-024-00630-2] [Citation(s) in RCA: 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: 11/18/2022] [Accepted: 06/26/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS), the most common endocrine disorder in premenopausal women, is associated with increased obesity, hyperandrogenism, and altered brown adipose tissue (BAT) thermogenesis. MicroRNAs play critical functions in brown adipocyte differentiation and maintenance. We aim to study the role of microRNA-21 (miR-21) in altered energy homeostasis and BAT thermogenesis in a PCOS mouse model of peripubertal androgen exposure. METHODS Three-week-old miR-21 knockout (miR21KO) or wild-type (WT) female mice were treated with dihydrotestosterone (DHT) or vehicle for 90 days. Body composition was determined by EchoMRI. Energy expenditure (EE), oxygen consumption (VO2), carbon dioxide production (VCO2), and respiratory exchange ratio (RER) were measured by indirect calorimetry. Androgen receptor (AR), and markers of adipogenesis, de novo lipogenesis, angiogenesis, extracellular matrix remodeling, and thermogenesis were quantified by RT-qPCR and/or Western-blot. RESULTS MiR-21 ablation attenuated DHT-mediated increase in body weight while having no effect on fat or BAT mass. MiR-21 ablation attenuated DHT-mediated BAT AR upregulation. MiR-21 ablation did not alter EE; however, miR21KO DHT-treated mice have reduced VO2, VCO2, and RER. MiR-21 ablation reversed DHT-mediated decrease in food intake and increase in sleep time. MiR-21 ablation decreased some adipogenesis (Adipoq, Pparγ, and Cebpβ) and extracellular matrix remodeling (Mmp-9 and Timp-1) markers expression in DHT-treated mice. MiR-21 ablation abolished DHT-mediated increases in thermogenesis markers Cpt1a and Cpt1b, while decreasing CIDE-A expression. CONCLUSIONS Our findings suggest that BAT miR-21 may play a role in regulating DHT-mediated thermogenic dysfunction in PCOS. Modulation of BAT miR-21 levels could be a novel therapeutic approach for the treatment of PCOS-associated metabolic derangements.
Collapse
Affiliation(s)
- Samar Rezq
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
| | - Alexandra M Huffman
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Jelina Basnet
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Amira E Alsemeh
- Department of Anatomy, Histology, and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Jussara M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Licy L Yanes Cardozo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Medicine, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Damian G Romero
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
3
|
Chand S, Tripathi AS, Dewani AP, Sheikh NWA. Molecular targets for management of diabetes: Remodelling of white adipose to brown adipose tissue. Life Sci 2024; 345:122607. [PMID: 38583857 DOI: 10.1016/j.lfs.2024.122607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/28/2024] [Accepted: 04/01/2024] [Indexed: 04/09/2024]
Abstract
Diabetes mellitus is a disorder characterised metabolic dysfunction that results in elevated glucose level in the bloodstream. Diabetes is of two types, type1 and type 2 diabetes. Obesity is considered as one of the major reasons intended for incidence of diabetes hence it turns out to be essential to study about the adipose tissue which is responsible for fat storage in body. Adipose tissues play significant role in maintaining the balance between energy stabilization and homeostasis. The three forms of adipose tissue are - White adipose tissue (WAT), Brown adipose tissue (BAT) and Beige adipose tissue (intermediate form). The amount of BAT gets reduced, and WAT starts to increase with the age. WAT when exposed to certain stimuli gets converted to BAT by the help of certain transcriptional regulators. The browning of WAT has been a matter of study to treat the metabolic disorders and to initiate the expenditure of energy. The three main regulators responsible for the browning of WAT are PRDM16, PPARγ and PGC-1α via various cellular and molecular mechanism. Presented review article includes the detailed elaborative aspect of genes and proteins involved in conversion of WAT to BAT.
Collapse
Affiliation(s)
- Shushmita Chand
- Amity Institute of Pharmacy, Amity University, Sector 125, Noida, Uttar Pradesh, India
| | - Alok Shiomurti Tripathi
- Department of Pharmacology, ERA College of Pharmacy, ERA University, Lucknow, Uttar Pradesh, India.
| | - Anil P Dewani
- Department of Pharmacology, P. Wadhwani College of Pharmacy, Yavatmal, Maharashtra, India
| | | |
Collapse
|
4
|
Zalaquett NG, Salameh E, Kim JM, Ghanbarian E, Tawk K, Abouzari M. The Dawn and Advancement of the Knowledge of the Genetics of Migraine. J Clin Med 2024; 13:2701. [PMID: 38731230 PMCID: PMC11084801 DOI: 10.3390/jcm13092701] [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: 03/20/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Background: Migraine is a prevalent episodic brain disorder known for recurrent attacks of unilateral headaches, accompanied by complaints of photophobia, phonophobia, nausea, and vomiting. Two main categories of migraine are migraine with aura (MA) and migraine without aura (MO). Main body: Early twin and population studies have shown a genetic basis for these disorders, and efforts have been invested since to discern the genes involved. Many techniques, including candidate-gene association studies, loci linkage studies, genome-wide association, and transcription studies, have been used for this goal. As a result, several genes were pinned with concurrent and conflicting data among studies. It is important to understand the evolution of techniques and their findings. Conclusions: This review provides a chronological understanding of the different techniques used from the dawn of migraine genetic investigations and the genes linked with the migraine subtypes.
Collapse
Affiliation(s)
- Nader G. Zalaquett
- Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Elio Salameh
- Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Jonathan M. Kim
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92697, USA
| | - Elham Ghanbarian
- Department of Neurology, University of California, Irvine, CA 92617, USA
| | - Karen Tawk
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92697, USA
| | - Mehdi Abouzari
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92697, USA
| |
Collapse
|
5
|
Kühnisch J, Theisen S, Dartsch J, Fritsche-Guenther R, Kirchner M, Obermayer B, Bauer A, Kahlert AK, Rothe M, Beule D, Heuser A, Mertins P, Kirwan JA, Berndt N, MacRae CA, Hubner N, Klaassen S. Prdm16 mutation determines sex-specific cardiac metabolism and identifies two novel cardiac metabolic regulators. Cardiovasc Res 2024; 119:2902-2916. [PMID: 37842925 PMCID: PMC10874277 DOI: 10.1093/cvr/cvad154] [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: 01/19/2023] [Revised: 07/04/2023] [Accepted: 07/18/2023] [Indexed: 10/17/2023] Open
Abstract
AIMS Mutation of the PRDM16 gene causes human dilated and non-compaction cardiomyopathy. The PRDM16 protein is a transcriptional regulator that affects cardiac development via Tbx5 and Hand1, thus regulating myocardial structure. The biallelic inactivation of Prdm16 induces severe cardiac dysfunction with post-natal lethality and hypertrophy in mice. The early pathological events that occur upon Prdm16 inactivation have not been explored. METHODS AND RESULTS This study performed in-depth pathophysiological and molecular analyses of male and female Prdm16csp1/wt mice that carry systemic, monoallelic Prdm16 gene inactivation. We systematically assessed early molecular changes through transcriptomics, proteomics, and metabolomics. Kinetic modelling of cardiac metabolism was performed in silico with CARDIOKIN. Prdm16csp1/wt mice are viable up to 8 months, develop hypoplastic hearts, and diminished systolic performance that is more pronounced in female mice. Prdm16csp1/wt cardiac tissue of both sexes showed reductions in metabolites associated with amino acid as well as glycerol metabolism, glycolysis, and the tricarboxylic acid cycle. Prdm16csp1/wt cardiac tissue revealed diminished glutathione (GSH) and increased inosine monophosphate (IMP) levels indicating oxidative stress and a dysregulated energetics, respectively. An accumulation of triacylglycerides exclusively in male Prdm16csp1/wt hearts suggests a sex-specific metabolic adaptation. Metabolic modelling using CARDIOKIN identified a reduction in fatty acid utilization in males as well as lower glucose utilization in female Prdm16csp1/wt cardiac tissue. On the level of transcripts and protein expression, Prdm16csp1/wt hearts demonstrate an up-regulation of pyridine nucleotide-disulphide oxidoreductase domain 2 (Pyroxd2) and the transcriptional regulator pre-B-cell leukaemia transcription factor interacting protein 1 (Pbxip1). The strongest concordant transcriptional up-regulation was detected for Prdm16 itself, probably through an autoregulatory mechanism. CONCLUSIONS Monoallelic, global Prdm16 mutation diminishes cardiac performance in Prdm16csp1/wt mice. Metabolic alterations and transcriptional dysregulation in Prdm16csp1/wt affect cardiac tissue. Female Prdm16csp1/wt mice develop a more pronounced phenotype, indicating sexual dimorphism at this early pathological window. This study suggests that metabolic dysregulation is an early event in the PRDM16 associated cardiac pathology.
Collapse
Affiliation(s)
- Jirko Kühnisch
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Lindenberger Weg 80, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Simon Theisen
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Lindenberger Weg 80, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Josephine Dartsch
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Lindenberger Weg 80, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Raphaela Fritsche-Guenther
- BIH Metabolomics Platform, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marieluise Kirchner
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Proteomics Platform, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Benedikt Obermayer
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Bauer
- BIH Metabolomics Platform, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anne-Karin Kahlert
- Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital of Schleswig-Holstein, Kiel, Germany
- DZHK German Center for Cardiovascular Research, partner site Hamburg/Kiel/Lübeck, Germany
- Institute of Immunology and Genetics, Kaiserslautern, Germany
| | | | - Dieter Beule
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Arnd Heuser
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Philipp Mertins
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Proteomics Platform, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jennifer A Kirwan
- BIH Metabolomics Platform, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nikolaus Berndt
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Computer-assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam—Rehbruecke (DIfE), Nuthetal, Germany
| | - Calum A MacRae
- Harvard Medical School and Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, USA
| | - Norbert Hubner
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Sabine Klaassen
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Lindenberger Weg 80, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Pediatric Cardiology, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| |
Collapse
|
6
|
Xu F, Jiang H, Li X, Pan J, Li H, Wang L, Zhang P, Chen J, Qiu S, Xie Y, Li Y, Zhang D, Dong Z. Discovery of PRDM16-Mediated TRPA1 Induction as the Mechanism for Low Tubulo-Interstitial Fibrosis in Diabetic Kidney Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306704. [PMID: 38072665 PMCID: PMC10870028 DOI: 10.1002/advs.202306704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/03/2023] [Indexed: 02/17/2024]
Abstract
The pathogenesis of Diabetic kidney disease(DKD) involves pathological changes in both tubulo-interstitium and the glomerulus. Surprisingly, tubulo-interstitial fibrosis (TIF), does not develop significantly until the late stage of DKD. Here, it is demonstrated that PR domain-containing 16 (PRDM16) is a key to the low level of TIF in DKD. In the experiments, PRDM16 is upregulated in high glucose-treated renal tubular cells, DKD mouse kidneys, and renal biopsy of human DKD patients via activation of NF-κB signal pathway. High glucose-induced expression of fibrotic proteins in renal tubular cells is suppressed by PRDM16. Mechanistically, PRDM16 bound to the promotor region of Transient receptor potential ankyrin 1 (TRPA1) to transactivate its expression and then suppressed MAPK (P38, ERK1/2) activation and downstream expression of TGF-β1. Knockout of PRDM16 from kidney proximal tubules in mice blocked TRPA1 expression and enhanced MAPK activation, TGF-β1 production, TIF development, and DKD progression, whereas knock-in of PRDM16 has opposite effects. In addition, overexpression of PRDM16 or its induction by formononetin ameliorated renal dysfunction and fibrosis in db/db diabetic mice. Finally, the above finding are detected in renal biopsies of DKD patients. Together, these results unveil PRDM16/TRPA1 as the mechanism responsible for the low level of TIF in the early stage of DKD by suppressing and TGF-β1 expression.
Collapse
Affiliation(s)
- Fang Xu
- Department of Emergency MedicineSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Emergency Medicine and Difficult Diseases InstituteSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Department of NephrologySecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
| | - Hongwei Jiang
- Department of EndocrinologyFirst Affiliated Hospital of Henan University of Science and TechnologyLuoyangHenan471000P. R. China
| | - Xiaozhou Li
- Department of Emergency MedicineSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Emergency Medicine and Difficult Diseases InstituteSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
| | - Jian Pan
- Department of Emergency MedicineSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Emergency Medicine and Difficult Diseases InstituteSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
| | - Huiling Li
- Department of OphthalmologyCentral South UniversityChangshaHunan410011P. R. China
| | - Luxiang Wang
- Department of Emergency MedicineSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Emergency Medicine and Difficult Diseases InstituteSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
| | - Pan Zhang
- Department of Emergency MedicineSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Emergency Medicine and Difficult Diseases InstituteSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Department of Epidemiology and Health StatisticsXiangya School of Public HealthCentral South UniversityChangshaHunan410011P. R. China
| | - Junxiang Chen
- Department of NephrologySecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
| | - Shuangfa Qiu
- Department of Emergency MedicineSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Emergency Medicine and Difficult Diseases InstituteSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
| | - Yuxin Xie
- Department of Emergency MedicineSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Emergency Medicine and Difficult Diseases InstituteSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
| | - Yijian Li
- Department of UrologySecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
| | - Dongshan Zhang
- Department of Emergency MedicineSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Emergency Medicine and Difficult Diseases InstituteSecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Department of NephrologySecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Department of EndocrinologyFirst Affiliated Hospital of Henan University of Science and TechnologyLuoyangHenan471000P. R. China
| | - Zheng Dong
- Department of NephrologySecond Xiangya HospitalCentral South UniversityChangshaHunan410011P. R. China
- Department of Cellular Biology and AnatomyMedical College of Georgia at Augusta UniversityAugustaGeorgia30906USA
| |
Collapse
|
7
|
Ramón A, Esteves A, Villadóniga C, Chalar C, Castro-Sowinski S. A general overview of the multifactorial adaptation to cold: biochemical mechanisms and strategies. Braz J Microbiol 2023; 54:2259-2287. [PMID: 37477802 PMCID: PMC10484896 DOI: 10.1007/s42770-023-01057-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023] Open
Abstract
Cold environments are more frequent than people think. They include deep oceans, cold lakes, snow, permafrost, sea ice, glaciers, cold soils, cold deserts, caves, areas at elevations greater than 3000 m, and also artificial refrigeration systems. These environments are inhabited by a diversity of eukaryotic and prokaryotic organisms that must adapt to the hard conditions imposed by cold. This adaptation is multifactorial and includes (i) sensing the cold, mainly through the modification of the liquid-crystalline membrane state, leading to the activation of a two-component system that transduce the signal; (ii) adapting the composition of membranes for proper functions mainly due to the production of double bonds in lipids, changes in hopanoid composition, and the inclusion of pigments; (iii) producing cold-adapted proteins, some of which show modifications in the composition of amino acids involved in stabilizing interactions and structural adaptations, e.g., enzymes with high catalytic efficiency; and (iv) producing ice-binding proteins and anti-freeze proteins, extracellular polysaccharides and compatible solutes that protect cells from intracellular and extracellular ice. However, organisms also respond by reprogramming their metabolism and specifically inducing cold-shock and cold-adaptation genes through strategies such as DNA supercoiling, distinctive signatures in promoter regions and/or the action of CSPs on mRNAs, among others. In this review, we describe the main findings about how organisms adapt to cold, with a focus in prokaryotes and linking the information with findings in eukaryotes.
Collapse
Affiliation(s)
- Ana Ramón
- Sección Bioquímica, Instituto de Biología, Facultad de Ciencias, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay
| | - Adriana Esteves
- Sección Bioquímica, Instituto de Biología, Facultad de Ciencias, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay
| | - Carolina Villadóniga
- Laboratorio de Biocatalizadores Y Sus Aplicaciones, Facultad de Ciencias, Instituto de Química Biológica, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay
| | - Cora Chalar
- Sección Bioquímica, Instituto de Biología, Facultad de Ciencias, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay
| | - Susana Castro-Sowinski
- Sección Bioquímica, Instituto de Biología, Facultad de Ciencias, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay.
- Laboratorio de Biocatalizadores Y Sus Aplicaciones, Facultad de Ciencias, Instituto de Química Biológica, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay.
| |
Collapse
|
8
|
Abstract
Brown adipose tissue (BAT) displays the unique capacity to generate heat through uncoupled oxidative phosphorylation that makes it a very attractive therapeutic target for cardiometabolic diseases. Here, we review BAT cellular metabolism, its regulation by the central nervous and endocrine systems and circulating metabolites, the plausible roles of this tissue in human thermoregulation, energy balance, and cardiometabolic disorders, and the current knowledge on its pharmacological stimulation in humans. The current definition and measurement of BAT in human studies relies almost exclusively on BAT glucose uptake from positron emission tomography with 18F-fluorodeoxiglucose, which can be dissociated from BAT thermogenic activity, as for example in insulin-resistant states. The most important energy substrate for BAT thermogenesis is its intracellular fatty acid content mobilized from sympathetic stimulation of intracellular triglyceride lipolysis. This lipolytic BAT response is intertwined with that of white adipose (WAT) and other metabolic tissues, and cannot be independently stimulated with the drugs tested thus far. BAT is an interesting and biologically plausible target that has yet to be fully and selectively activated to increase the body's thermogenic response and shift energy balance. The field of human BAT research is in need of methods able to directly, specifically, and reliably measure BAT thermogenic capacity while also tracking the related thermogenic responses in WAT and other tissues. Until this is achieved, uncertainty will remain about the role played by this fascinating tissue in human cardiometabolic diseases.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | | | - Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec City, Quebec, G1V 4G5, Canada
| |
Collapse
|
9
|
Rahman MS, Lee Y, Park DS, Kim YS. Bifidobacterium bifidum DS0908 and Bifidobacterium longum DS0950 Culture-Supernatants Ameliorate Obesity-Related Characteristics in Mice with High-Fat Diet-Induced Obesity. J Microbiol Biotechnol 2023; 33:96-105. [PMID: 36457182 PMCID: PMC9899789 DOI: 10.4014/jmb.2210.10046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022]
Abstract
Probiotic supplements have promising therapeutic effects on chronic diseases. In this study, we demonstrated the anti-obesity effects of two potential probiotics, Bifidobacterium bifidum DS0908 (DS0908) and Bifidobacterium longum DS0950 (DS0950). Treatment with DS0908 and DS0950 postbiotics significantly induced the expression of the brown adipocyte-specific markers UCP1, PPARγ, PGC1α, PRDM16 and beige adipocyte-specific markers CD137, FGF21, P2RX5, and COX2 in C3H10T1/2 mesenchymal stem cells (MSCs). In mice with high-fat diet (HFD)-induced obesity, both potential probiotics and postbiotics noticeably reduced body weight and epididymal fat accumulation without affecting food intake. DS0908 and DS0950 also improved insulin sensitivity and glucose use in mice with HFD-induced obesity. In addition, DS0908 and DS0950 improved the plasma lipid profile, proved by reduced triglyceride, low-density lipoprotein, and cholesterol levels. Furthermore, DS0908 and DS0950 improved mitochondrial respiratory function, confirmed by the high expression of oxidative phosphorylation proteins, during thermogenesis induction in the visceral and epididymal fat in mice with HFD-induced obesity. Notably, the physiological and metabolic changes were more significant after treatment with potential probiotic culture-supernatants than those with the bacterial pellet. Finally, gene knockdown and co-treatment with inhibitor-mediated mechanistic analyses showed that both DS0908 and DS0950 exerted anti-obesity-related effects via the PKA/p38 MAPK signaling activation in C3H10T1/2 MSCs. Our observations suggest that DS0908 and DS0950 could potentially alleviate obesity as dietary supplements.
Collapse
Affiliation(s)
- M. Shamim Rahman
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea,Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Youri Lee
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea,Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Doo-Sang Park
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Yong-Sik Kim
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea,Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea,Corresponding author Phone: +82-41-570-2413 Fax: +82-41-575-2412 E-mail:
| |
Collapse
|
10
|
Armani A, Feraco A, Camajani E, Gorini S, Lombardo M, Caprio M. Nutraceuticals in Brown Adipose Tissue Activation. Cells 2022; 11:cells11243996. [PMID: 36552762 PMCID: PMC9776638 DOI: 10.3390/cells11243996] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Obesity and its associated comorbidities have become pandemic, and challenge the global healthcare system. Lifestyle changes, nutritional interventions and phamaceuticals should be differently combined in a personalized strategy to tackle such a public health burden. Altered brown adipose tissue (BAT) function contributes to the pathophysiology of obesity and glucose metabolism dysfunctions. BAT thermogenic activity burns glucose and fatty acids to produce heat through uncoupled respiration, and can dissipate the excessive calorie intake, reduce glycemia and circulate fatty acids released from white adipose tissue. Thus, BAT activity is expected to contribute to whole body energy homeostasis and protect against obesity, diabetes and alterations in lipid profile. To date, pharmacological therapies aimed at activating brown fat have failed in clinical trials, due to cardiovascular side effects or scarce efficacy. On the other hand, several studies have identified plant-derived chemical compounds capable of stimulating BAT thermogenesis in animal models, suggesting the translational applications of dietary supplements to fight adipose tissue dysfunctions. This review describes several nutraceuticals with thermogenic properties and provides indications, at a molecular level, of the regulation of the adipocyte thermogenesis by the mentioned phytochemicals.
Collapse
Affiliation(s)
- Andrea Armani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
- Correspondence:
| | - Alessandra Feraco
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Elisabetta Camajani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Stefania Gorini
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Mauro Lombardo
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Massimiliano Caprio
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| |
Collapse
|
11
|
Nikolic M, Novakovic J, Ramenskaya G, Kokorekin V, Jeremic N, Jakovljevic V. Cooling down with Entresto. Can sacubitril/valsartan combination enhance browning more than coldness? Diabetol Metab Syndr 2022; 14:175. [PMID: 36419097 PMCID: PMC9686067 DOI: 10.1186/s13098-022-00944-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND It is a growing importance to induce a new treatment approach to encourage weight loss but also to improve maintenance of lost weight. It has been shown that promotion of brown adipose tissue (BAT) function or acquisition of BAT characteristics in white adipose tissue (terms referred as "browning") can be protective against obesity. MAIN TEXT Amongst numerous established environmental influences on BAT activity, cold exposure is the best interested technique due to its not only effects on of BAT depots in proliferation process but also de novo differentiation of precursor cells via β-adrenergic receptor activation. A novel combination drug, sacubitril/valsartan, has been shown to be more efficient in reducing cardiovascular events and heart failure readmission compared to conventional therapy. Also, this combination of drugs increases the postprandial lipid oxidation contributing to energy expenditure, promotes lipolysis in adipocytes and reduces body weight. To date, there is no research examining potential of combined sacubitril/valsartan use to promote browning or mechanisms in the basis of this thermogenic process. CONCLUSION Due to the pronounced effects of cold and sacubitril/valsartan treatment on function and metabolism of BAT, the primary goal of further research should focused on investigation of the synergistic effects of the sacubitril/valsartan treatment at low temperature environmental conditions.
Collapse
Affiliation(s)
- Marina Nikolic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Jovana Novakovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | | | - Nevena Jeremic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
- First Moscow State Medical University IM Sechenov, Moscow, Russia.
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Human Pathology, First Moscow State Medical University IM Sechenov, Moscow, Russia
| |
Collapse
|
12
|
Ebeid M, Barnas K, Zhang H, Yaghmour A, Noreikaite G, Bjork BC. PRDM16 expression and function in mammalian cochlear development. Dev Dyn 2022; 251:1666-1683. [PMID: 35451126 PMCID: PMC9790675 DOI: 10.1002/dvdy.480] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND PR domain containing 16 (PRDM16) is a key transcriptional regulator in the development of craniofacial, adipose, and neural tissues. Our lab identified PRDM16 expression in the epithelial cells of the Kölliker's organ (KO) that starts at ~E13.5 and is maintained until KO disappearance. A transgenic mouse model that carries a gene trap null allele of Prdm16 (Prdm16cGT ) was used to characterize the impact of Prdm16 loss on cochlear development. RESULTS At P0 Prdm16cGT null cochlea exhibited hypoplastic KO, shortened cochlear duct, increased density of hair cells (HCs) and supporting cells (SCs) in the apical turn as well as multiple isolated ectopic HCs within the KO domain. KO epithelial cells proliferation rate was reduced in the apical turn of the developing Prdm16cGT null cochlea vs controls. Bulk RNA sequencing of cochlear duct cells at E14.5 followed by quantitative real time PCR and mRNA Fluorescence in-situ hybridization (FISH) validation identified differentially expressed genes in Prdm16cGT null vs littermate control cochleae. Upregulated genes at E14.5 included Fgf20, as well as several Notch pathway genes (Lfng, Hes1, and Jag1). CONCLUSIONS This study characterizes Prdm16 expression during cochlear development and establishes its requirement for KO development.
Collapse
Affiliation(s)
- Michael Ebeid
- College of Graduate Studies, Midwestern UniversityDowners GroveIllinoisUSA,Department of AnatomyMidwestern UniversityDowners GroveIllinoisUSA,Chicago College of Osteopathic MedicineMidwestern UniversityDowners GroveIllinoisUSA
| | - Kathy Barnas
- Biomedical Sciences ProgramMidwestern UniversityDowners GroveIllinoisUSA
| | - Hongji Zhang
- Department of AnatomyMidwestern UniversityDowners GroveIllinoisUSA
| | - Amal Yaghmour
- Biomedical Sciences ProgramMidwestern UniversityDowners GroveIllinoisUSA
| | - Gabriele Noreikaite
- Chicago College of Osteopathic MedicineMidwestern UniversityDowners GroveIllinoisUSA
| | - Bryan C. Bjork
- College of Graduate Studies, Midwestern UniversityDowners GroveIllinoisUSA
| |
Collapse
|
13
|
Machado SA, Pasquarelli-do-Nascimento G, da Silva DS, Farias GR, de Oliveira Santos I, Baptista LB, Magalhães KG. Browning of the white adipose tissue regulation: new insights into nutritional and metabolic relevance in health and diseases. Nutr Metab (Lond) 2022; 19:61. [PMID: 36068578 PMCID: PMC9446768 DOI: 10.1186/s12986-022-00694-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/19/2022] [Indexed: 12/11/2022] Open
Abstract
Adipose tissues are dynamic tissues that play crucial physiological roles in maintaining health and homeostasis. Although white adipose tissue and brown adipose tissue are currently considered key endocrine organs, they differ functionally and morphologically. The existence of the beige or brite adipocytes, cells displaying intermediary characteristics between white and brown adipocytes, illustrates the plastic nature of the adipose tissue. These cells are generated through white adipose tissue browning, a process associated with augmented non-shivering thermogenesis and metabolic capacity. This process involves the upregulation of the uncoupling protein 1, a molecule that uncouples the respiratory chain from Adenosine triphosphate synthesis, producing heat. β-3 adrenergic receptor system is one important mediator of white adipose tissue browning, during cold exposure. Surprisingly, hyperthermia may also induce beige activation and white adipose tissue beiging. Physical exercising copes with increased levels of specific molecules, including Beta-Aminoisobutyric acid, irisin, and Fibroblast growth factor 21 (FGF21), which induce adipose tissue browning. FGF21 is a stress-responsive hormone that interacts with beta-klotho. The central roles played by hormones in the browning process highlight the relevance of the individual lifestyle, including circadian rhythm and diet. Circadian rhythm involves the sleep-wake cycle and is regulated by melatonin, a hormone associated with UCP1 level upregulation. In contrast to the pro-inflammatory and adipose tissue disrupting effects of the western diet, specific food items, including capsaicin and n-3 polyunsaturated fatty acids, and dietary interventions such as calorie restriction and intermittent fasting, favor white adipose tissue browning and metabolic efficiency. The intestinal microbiome has also been pictured as a key factor in regulating white tissue browning, as it modulates bile acid levels, important molecules for the thermogenic program activation. During embryogenesis, in which adipose tissue formation is affected by Bone morphogenetic proteins that regulate gene expression, the stimuli herein discussed influence an orchestra of gene expression regulators, including a plethora of transcription factors, and chromatin remodeling enzymes, and non-coding RNAs. Considering the detrimental effects of adipose tissue browning and the disparities between adipose tissue characteristics in mice and humans, further efforts will benefit a better understanding of adipose tissue plasticity biology and its applicability to managing the overwhelming burden of several chronic diseases.
Collapse
Affiliation(s)
- Sabrina Azevedo Machado
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasília, DF, Brazil
| | | | - Debora Santos da Silva
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasília, DF, Brazil
| | - Gabriel Ribeiro Farias
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasília, DF, Brazil
| | - Igor de Oliveira Santos
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasília, DF, Brazil
| | - Luana Borges Baptista
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasília, DF, Brazil
| | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasília, DF, Brazil.
| |
Collapse
|
14
|
Fernandez NC, Shinoda K. The Role of B Lymphocyte Subsets in Adipose Tissue Development, Metabolism, and Aging. Compr Physiol 2022; 12:4133-4145. [PMID: 35950657 DOI: 10.1002/cphy.c220006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adipose tissue contains resident B lymphocytes (B cells) with varying immune functions and mechanisms, depending on the adipose depot type and location. The heterogeneity of B cells and their functions affect the immunometabolism of the adipose tissue in aging and age-associated metabolic disorders. B cells exist in categorizations of subsets that have developmental or phenotypic differences with varying functionalities. Subsets can be categorized as either protective or pathogenic depending on their secretion profile or involvement in metabolic maintenance. In this article, we summarized recent finding on the B cell heterogeneity and discuss how we can utilize our current knowledge of adipose resident B lymphocytes for potential treatment for age-associated metabolic disorders. © 2022 American Physiological Society. Compr Physiol 12: 1-13, 2022.
Collapse
Affiliation(s)
- Nicole C Fernandez
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kosaku Shinoda
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Division of Endocrinology & Diabetes, Albert Einstein College of Medicine, Bronx, New York, USA
- Fleischer Institute for Diabetes and Metabolism, Bronx, New York, USA
| |
Collapse
|
15
|
Scheel AK, Espelage L, Chadt A. Many Ways to Rome: Exercise, Cold Exposure and Diet-Do They All Affect BAT Activation and WAT Browning in the Same Manner? Int J Mol Sci 2022; 23:ijms23094759. [PMID: 35563150 PMCID: PMC9103087 DOI: 10.3390/ijms23094759] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 02/08/2023] Open
Abstract
The discovery of functional brown adipose tissue (BAT) in adult humans and the possibility to recruit beige cells with high thermogenic potential within white adipose tissue (WAT) depots opened the field for new strategies to combat obesity and its associated comorbidities. Exercise training as well as cold exposure and dietary components are associated with the enhanced accumulation of metabolically-active beige adipocytes and BAT activation. Both activated beige and brown adipocytes increase their metabolic rate by utilizing lipids to generate heat via non-shivering thermogenesis, which is dependent on uncoupling protein 1 (UCP1) in the inner mitochondrial membrane. Non-shivering thermogenesis elevates energy expenditure and promotes a negative energy balance, which may ameliorate metabolic complications of obesity and Type 2 Diabetes Mellitus (T2DM) such as insulin resistance (IR) in skeletal muscle and adipose tissue. Despite the recent advances in pharmacological approaches to reduce obesity and IR by inducing non-shivering thermogenesis in BAT and WAT, the administered pharmacological compounds are often associated with unwanted side effects. Therefore, lifestyle interventions such as exercise, cold exposure, and/or specified dietary regimens present promising anchor points for future disease prevention and treatment of obesity and T2DM. The exact mechanisms where exercise, cold exposure, dietary interventions, and pharmacological treatments converge or rather diverge in their specific impact on BAT activation or WAT browning are difficult to determine. In the past, many reviews have demonstrated the mechanistic principles of exercise- and/or cold-induced BAT activation and WAT browning. In this review, we aim to summarize not only the current state of knowledge on the various mechanistic principles of diverse external stimuli on BAT activation and WAT browning, but also present their translational potential in future clinical applications.
Collapse
Affiliation(s)
- Anna K. Scheel
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Medical Faculty, Düsseldorf, Auf’m Hennekamp 65, 40225 Duesseldorf, Germany; (A.K.S.); (L.E.)
- German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, 85764 München, Germany
| | - Lena Espelage
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Medical Faculty, Düsseldorf, Auf’m Hennekamp 65, 40225 Duesseldorf, Germany; (A.K.S.); (L.E.)
- German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, 85764 München, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Medical Faculty, Düsseldorf, Auf’m Hennekamp 65, 40225 Duesseldorf, Germany; (A.K.S.); (L.E.)
- German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, 85764 München, Germany
- Correspondence: ; Tel./Fax: +49-211-3382-577/430
| |
Collapse
|
16
|
Bast-Habersbrunner A, Kiefer C, Weber P, Fromme T, Schießl A, Schwalie PC, Deplancke B, Li Y, Klingenspor M. LncRNA Ctcflos orchestrates transcription and alternative splicing in thermogenic adipogenesis. EMBO Rep 2021; 22:e51289. [PMID: 34056831 PMCID: PMC8256291 DOI: 10.15252/embr.202051289] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
The recruitment of thermogenic brite adipocytes within white adipose tissue attenuates obesity and metabolic comorbidities, arousing interest in understanding the underlying regulatory mechanisms. The molecular network of brite adipogenesis, however, remains largely unresolved. In this light, long noncoding RNAs (lncRNAs) emerged as a versatile class of modulators that control many steps within the differentiation machinery. Leveraging the naturally varying propensities of different inbred mouse strains for white adipose tissue browning, we identify the nuclear lncRNA Ctcflos as a pivotal orchestrator of thermogenic gene expression during brite adipocyte differentiation. Mechanistically, Ctcflos acts as a pleiotropic regulator, being essential for the transcriptional recruitment of the early core thermogenic regulatory program and the modulation of alternative splicing to drive brite adipogenesis. This is showcased by Ctcflos‐regulated gene transcription and splicing of the key browning factor Prdm16 toward the isoform that is specific for the thermogenic gene program. Conclusively, our findings emphasize the mechanistic versatility of lncRNAs acting at several independent levels of gene expression for effective regulation of key differentiation factors to direct cell fate and function.
Collapse
Affiliation(s)
- Andrea Bast-Habersbrunner
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Christoph Kiefer
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Peter Weber
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Tobias Fromme
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Anna Schießl
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Petra C Schwalie
- School of Life Sciences, EPFL and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Bart Deplancke
- School of Life Sciences, EPFL and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Yongguo Li
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
| |
Collapse
|
17
|
Thermogenic adipocytes: lineage, function and therapeutic potential. Biochem J 2020; 477:2071-2093. [PMID: 32539124 PMCID: PMC7293110 DOI: 10.1042/bcj20200298] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022]
Abstract
Metabolic inflexibility, defined as the inability to respond or adapt to metabolic demand, is now recognised as a driving factor behind many pathologies associated with obesity and the metabolic syndrome. Adipose tissue plays a pivotal role in the ability of an organism to sense, adapt to and counteract environmental changes. It provides a buffer in times of nutrient excess, a fuel reserve during starvation and the ability to resist cold-stress through non-shivering thermogenesis. Recent advances in single-cell RNA sequencing combined with lineage tracing, transcriptomic and proteomic analyses have identified novel adipocyte progenitors that give rise to specialised adipocytes with diverse functions, some of which have the potential to be exploited therapeutically. This review will highlight the common and distinct functions of well-known adipocyte populations with respect to their lineage and plasticity, as well as introducing the most recent members of the adipocyte family and their roles in whole organism energy homeostasis. Finally, this article will outline some of the more preliminary findings from large data sets generated by single-cell transcriptomics of mouse and human adipose tissue and their implications for the field, both for discovery and for therapy.
Collapse
|
18
|
The Intricate Role of p53 in Adipocyte Differentiation and Function. Cells 2020; 9:cells9122621. [PMID: 33297294 PMCID: PMC7762213 DOI: 10.3390/cells9122621] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022] Open
Abstract
For more than three decades, numerous studies have demonstrated the function of p53 in cell cycle, cellular senescence, autophagy, apoptosis, and metabolism. Among diverse functions, the essential role of p53 is to maintain cellular homeostatic response to stress by regulating proliferation and apoptosis. Recently, adipocytes have been studied with increasing intensity owing to the increased prevalence of metabolic diseases posing a serious public health concern and because metabolic dysfunction can directly induce tumorigenesis. The prevalence of metabolic diseases has steadily increased worldwide, and a growing interest in these diseases has led to the focus on the role of p53 in metabolism and adipocyte differentiation with or without metabolic stress. However, our collective understanding of the direct role of p53 in adipocyte differentiation and function remains insufficient. Therefore, this review focuses on the newly discovered roles of p53 in adipocyte differentiation and function.
Collapse
|
19
|
Benlebna M, Balas L, Bonafos B, Pessemesse L, Fouret G, Vigor C, Gaillet S, Grober J, Bernex F, Landrier JF, Kuda O, Durand T, Coudray C, Casas F, Feillet-Coudray C. Long-term intake of 9-PAHPA or 9-OAHPA modulates favorably the basal metabolism and exerts an insulin sensitizing effect in obesogenic diet-fed mice. Eur J Nutr 2020; 60:2013-2027. [PMID: 32989473 DOI: 10.1007/s00394-020-02391-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Fatty acid esters of hydroxy fatty acids (FAHFAs) are a large family of endogenous bioactive lipids. To date, most of the studied FAHFAs are branched regioisomers of Palmitic Acid Hydroxyl Stearic Acid (PAHSA) that were reported to possess anti-diabetic and anti-inflammatory activity in humans and rodents. Recently, we have demonstrated that 9-PAHPA or 9-OAHPA intake increased basal metabolism and enhanced insulin sensitivity in healthy control diet-fed mice but induced liver damage in some mice. The present work aims to explore whether a long-term intake of 9-PAHPA or 9-OAHPA may have similar effects in obesogenic diet-fed mice. METHODS C57Bl6 mice were fed with a control or high fat-high sugar (HFHS) diets for 12 weeks. The HFHS diet was supplemented or not with 9-PAHPA or 9-OAHPA. Whole-body metabolism was explored. Glucose and lipid metabolism as well as mitochondrial activity and oxidative stress status were analyzed. RESULTS As expected, the intake of HFHS diet led to obesity and lower insulin sensitivity with minor effects on liver parameters. The long-term intake of 9-PAHPA or 9-OAHPA modulated favorably the basal metabolism and improved insulin sensitivity as measured by insulin tolerance test. On the contrary to what we have reported previously in healthy mice, no marked effect for these FAHFAs was observed on liver metabolism of obese diabetic mice. CONCLUSION This study indicates that both 9-PAHPA and 9-OAHPA may have interesting insulin-sensitizing effects in obese mice with lower insulin sensitivity.
Collapse
Affiliation(s)
| | - Laurence Balas
- Institut Des Biomolécules Max Mousseron (IBMM), Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | | | | | | | - Claire Vigor
- Institut Des Biomolécules Max Mousseron (IBMM), Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | | | - Jacques Grober
- LNC UMR1231, INSERM, Univ Bourgogne Franche-Comté, Agrosup Dijon, LipSTIC LabEx, Dijon, France
| | - Florence Bernex
- INSERM, U1194, Network of Experimental Histology, BioCampus, CNRS, UMS3426, Montpellier, France
| | | | - Ondrej Kuda
- Department of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Thierry Durand
- Institut Des Biomolécules Max Mousseron (IBMM), Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | | | | | | |
Collapse
|
20
|
Hussain MF, Roesler A, Kazak L. Regulation of adipocyte thermogenesis: mechanisms controlling obesity. FEBS J 2020; 287:3370-3385. [PMID: 32301220 DOI: 10.1111/febs.15331] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/26/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Adipocyte biology has been intensely researched in recent years due to the emergence of obesity as a serious global health concern and because of the realization that adipose tissue is more than simply a cell type that stores and releases lipids. The plasticity of adipose tissues, to rapidly adapt to altered physiological states of energy demand, is under neuronal and endocrine control. The capacity for white adipocytes to store chemical energy in lipid droplets is key for protecting other organs from the toxic effects of ectopic lipid deposition. In contrast, thermogenic (brown and beige) adipocytes combust macronutrients to generate heat. The thermogenic activity of adipocytes allows them to protect themselves and other tissues from lipid overaccumulation. Advances in brown fat biology have uncovered key molecular players involved in adipocyte determination, differentiation, and thermogenic activation. It is now, well appreciated that three distinct adipocyte types exist: white, beige, and brown. Moreover, functional differences are present within adipocyte subtypes located in anatomically distinct locations. Adding to this complexity is the recent realization from single-cell sequencing studies that adipocyte progenitors are also heterogeneous. Understanding the molecular details of how to increase the number of thermogenic fat cells and their activation may delineate some of the pathophysiological basis of obesity and obesity-related diseases. Here, we review recent advances that have extended our understanding of the central role that adipose tissue plays in energy balance and the mechanisms that control their amount and function.
Collapse
Affiliation(s)
- Mohammed Faiz Hussain
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Anna Roesler
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Lawrence Kazak
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| |
Collapse
|
21
|
Zhu R, Wei J, Liu H, Liu C, Wang L, Chen B, Li L, Jia Q, Tian Y, Li R, Zhao D, Mo F, Li Y, Gao S, Wang XD, Zhang D. Lycopene attenuates body weight gain through induction of browning via regulation of peroxisome proliferator-activated receptor γ in high-fat diet-induced obese mice. J Nutr Biochem 2020; 78:108335. [DOI: 10.1016/j.jnutbio.2019.108335] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 12/03/2019] [Accepted: 12/20/2019] [Indexed: 12/27/2022]
|
22
|
di Somma M, Vliora M, Grillo E, Castro B, Dakou E, Schaafsma W, Vanparijs J, Corsini M, Ravelli C, Sakellariou E, Mitola S. Role of VEGFs in metabolic disorders. Angiogenesis 2019; 23:119-130. [PMID: 31853841 DOI: 10.1007/s10456-019-09700-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023]
Abstract
Obesity and metabolic disorders are important public health problems. In this review, the role of vasculature network and VEGF in the adipose tissue maintenance and supplementation is discussed. Angiogenesis is a key process implicated in regulation of tissues homeostasis. Dysregulation of new blood vessels formation may be crucial and contribute to the onset of several pathological conditions, including metabolic syndrome-associated disorders. Adipose tissue homeostasis is fine regulated by vascular network. Vessels support adipose structure. Vasculature modulates the balance between positive and negative regulator factors. In white adipose tissue, vascular endothelial growth factor (VEGF) controls the metabolic activities of adipocytes promoting the trans-differentiation from white to beige phenotype. Trans-differentiation results in an increase of energy consumption. VEGF exerts an opposite effect on brown adipose tissue, where VEGF increases oxygen supply and improves energy expenditure inducing the whitening of adipocytes.
Collapse
Affiliation(s)
- M di Somma
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - M Vliora
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece
| | - E Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - B Castro
- Histocell, S.L.Parque Tecnológico 801A, 2º, 48160, Derio, Bizkaia, Spain
| | - E Dakou
- Laboratory of Cell Genetics, Department of Biology, Faculty of Science and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - W Schaafsma
- Histocell, S.L.Parque Tecnológico 801A, 2º, 48160, Derio, Bizkaia, Spain
| | - J Vanparijs
- Laboratory of Cell Genetics, Department of Biology, Faculty of Science and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - M Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - C Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - E Sakellariou
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece
| | - S Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
23
|
Wang W, Ishibashi J, Trefely S, Shao M, Cowan AJ, Sakers A, Lim HW, O'Connor S, Doan MT, Cohen P, Baur JA, King MT, Veech RL, Won KJ, Rabinowitz JD, Snyder NW, Gupta RK, Seale P. A PRDM16-Driven Metabolic Signal from Adipocytes Regulates Precursor Cell Fate. Cell Metab 2019; 30:174-189.e5. [PMID: 31155495 PMCID: PMC6836679 DOI: 10.1016/j.cmet.2019.05.005] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 03/28/2019] [Accepted: 05/01/2019] [Indexed: 12/18/2022]
Abstract
The precursor cells for metabolically beneficial beige adipocytes can alternatively become fibrogenic and contribute to adipose fibrosis. We found that cold exposure or β3-adrenergic agonist treatment of mice decreased the fibrogenic profile of precursor cells and stimulated beige adipocyte differentiation. This fibrogenic-to-adipogenic transition was impaired in aged animals, correlating with reduced adipocyte expression of the transcription factor PRDM16. Genetic loss of Prdm16 mimicked the effect of aging in promoting fibrosis, whereas increasing PRDM16 in aged mice decreased fibrosis and restored beige adipose development. PRDM16-expressing adipose cells secreted the metabolite β-hydroxybutyrate (BHB), which blocked precursor fibrogenesis and facilitated beige adipogenesis. BHB catabolism in precursor cells, mediated by BDH1, was required for beige fat differentiation in vivo. Finally, dietary BHB supplementation in aged animals reduced adipose fibrosis and promoted beige fat formation. Together, our results demonstrate that adipocytes secrete a metabolite signal that controls beige fat remodeling.
Collapse
Affiliation(s)
- Wenshan Wang
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeff Ishibashi
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sophie Trefely
- Department of Cancer Biology, University of Pennsylvania Philadelphia, PA, USA; AJ Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
| | - Mengle Shao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexis J Cowan
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ, 08544 USA
| | - Alexander Sakers
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Hee-Woong Lim
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Genetics Department, University of Pennsylvania, Philadelphia, PA, USA
| | - Sean O'Connor
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Mary T Doan
- AJ Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Joseph A Baur
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M Todd King
- Laboratory of Metabolic Control, NIH/NIAAA, Rockville, MD, USA
| | - Richard L Veech
- Laboratory of Metabolic Control, NIH/NIAAA, Rockville, MD, USA
| | - Kyoung-Jae Won
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Genetics Department, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ, 08544 USA
| | | | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Presby DM, Jackman MR, Rudolph MC, Sherk VD, Foright RM, Houck JA, Johnson GC, Orlicky DJ, Melanson EL, Higgins JA, MacLean PS. Compensation for cold-induced thermogenesis during weight loss maintenance and regain. Am J Physiol Endocrinol Metab 2019; 316:E977-E986. [PMID: 30912962 PMCID: PMC6580173 DOI: 10.1152/ajpendo.00543.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/19/2019] [Accepted: 03/19/2019] [Indexed: 11/22/2022]
Abstract
Prevalence of obesity is exacerbated by low rates of successful long-term weight loss maintenance (WLM). In part, relapse from WLM to obesity is due to a reduction in energy expenditure (EE) that persists throughout WLM and relapse. Thus, interventions that increase EE might facilitate WLM. In obese mice that were calorically restricted to reduce body weight by ~20%, we manipulated EE throughout WLM and early relapse using intermittent cold exposure (ICE; 4°C, 90 min/day, 5 days/wk, within the last 3 h of the light cycle). EE, energy intake, and spontaneous physical activity were measured during the obese, WLM, and relapse phases. During WLM and relapse, the ICE group expended more energy during the light cycle because of cold exposure but expended less energy in the dark cycle, which led to no overall difference in total daily EE. The compensation in EE appeared to be mediated by activity, whereby the ICE group was more active during the light cycle because of cold exposure but less active during the dark cycle, which led to no overall effect on total daily activity during WLM and relapse. In brown adipose tissue of relapsing mice, the ICE group had greater mRNA expression of Dio2 and protein expression of UCP1 but lower mRNA expression of Prdm16. In summary, these findings indicate that despite robust increases in EE during cold exposures, ICE is unable to alter total daily EE during WLM or early relapse, likely due to compensatory behaviors in activity.
Collapse
Affiliation(s)
- David M Presby
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Matthew R Jackman
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Michael C Rudolph
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Vanessa D Sherk
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Rebecca M Foright
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Julie A Houck
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Ginger C Johnson
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - David J Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Edward L Melanson
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus , Aurora, Colorado
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Janine A Higgins
- Department of Pediatrics, Section of Endocrinology, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Paul S MacLean
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| |
Collapse
|
25
|
Alcalá M, Calderon-Dominguez M, Serra D, Herrero L, Viana M. Mechanisms of Impaired Brown Adipose Tissue Recruitment in Obesity. Front Physiol 2019; 10:94. [PMID: 30814954 PMCID: PMC6381290 DOI: 10.3389/fphys.2019.00094] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/25/2019] [Indexed: 12/18/2022] Open
Abstract
Brown adipose tissue (BAT) dissipates energy to produce heat. Thus, it has the potential to regulate body temperature by thermogenesis. For the last decade, BAT has been in the spotlight due to its rediscovery in adult humans. This is evidenced by over a hundred clinical trials that are currently registered to target BAT as a therapeutic tool in the treatment of metabolic diseases, such as obesity or diabetes. The goal of most of these trials is to activate the BAT thermogenic program via several approaches such as adrenergic stimulation, natriuretic peptides, retinoids, capsinoids, thyroid hormones, or glucocorticoids. However, the impact of BAT activation on total body energy consumption and the potential effect on weight loss is still limited. Other studies have focused on increasing the mass of thermogenic BAT. This can be relevant in obesity, where the activity and abundance of BAT have been shown to be drastically reduced. The aim of this review is to describe pathological processes associated with obesity that may influence the correct differentiation of BAT, such as catecholamine resistance, inflammation, oxidative stress, and endoplasmic reticulum stress. This will shed light on the thermogenic potential of BAT as a therapeutic approach to target obesity-induced metabolic diseases.
Collapse
Affiliation(s)
- Martín Alcalá
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - María Calderon-Dominguez
- Department of Biochemistry and Physiology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Viana
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
26
|
Li AN, Chen JJ, Li QQ, Zeng GY, Chen QY, Chen JL, Liao ZM, Jin P, Wang KS, Yang ZC. Alpha-glucosidase inhibitor 1-Deoxynojirimycin promotes beige remodeling of 3T3-L1 preadipocytes via activating AMPK. Biochem Biophys Res Commun 2019; 509:1001-1007. [DOI: 10.1016/j.bbrc.2019.01.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/05/2019] [Indexed: 12/11/2022]
|
27
|
Marinovic MP, Campeiro JD, Lima SC, Rocha AL, Nering MB, Oliveira EB, Mori MA, Hayashi MAF. Crotamine induces browning of adipose tissue and increases energy expenditure in mice. Sci Rep 2018; 8:5057. [PMID: 29567992 PMCID: PMC5864908 DOI: 10.1038/s41598-018-22988-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/01/2018] [Indexed: 12/31/2022] Open
Abstract
Crotamine, originally isolated from rattlesnake venom, has been extensively studied due to its pleiotropic biological properties, and special attention has been paid to its antitumor activity. However, long-term treatment with crotamine was accompanied by a reduction in animal body weight gain and by increases in glucose tolerance. As cancer is commonly associated with cachexia, to preclude the possible cancer cachexia-like effect of crotamine, herein this polypeptide was administered in healthy wild-type C57/BL6 mice by the oral route daily, for 21 days. Reduced body weight gain, in addition to decreased white adipose tissue (WAT) and increased brown adipose tissue (BAT) mass were observed in healthy animals in the absence of tumor. In addition, we observed improved glucose tolerance and increased insulin sensitivity, accompanied by a reduction of plasma lipid levels and decreased levels of biomarkers of liver damage and kidney disfunctions. Importantly, long-term treatment with crotamine increased the basal metabolic rate in vivo, which was consistent with the increased expression of thermogenic markers in BAT and WAT. Interestingly, cultured brown adipocyte cells induced to differentiation in the presence of crotamine also showed increases in some of these markers and in lipid droplets number and size, indicating increased brown adipocyte maturation.
Collapse
Affiliation(s)
- Marcelo P Marinovic
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Joana D Campeiro
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Sunamita C Lima
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Andrea L Rocha
- Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil
| | - Marcela B Nering
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Eduardo B Oliveira
- Departamento de Bioquímica e Imunologia, Universidade de São Paulo (USP-RP), Ribeirão Preto, Brazil
| | - Marcelo A Mori
- Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil.,Departamento de Bioquímica e Biologia Tecidual, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Mirian A F Hayashi
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
| |
Collapse
|
28
|
Ma X, Wang D, Zhao W, Xu L. Deciphering the Roles of PPARγ in Adipocytes via Dynamic Change of Transcription Complex. Front Endocrinol (Lausanne) 2018; 9:473. [PMID: 30186237 PMCID: PMC6110914 DOI: 10.3389/fendo.2018.00473] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ), a ligand-dependent transcription factor highly expressed in adipocytes, is a master regulator of adipogenesis and lipid storage, a central player in thermogenesis and an active modulator of lipid metabolism and insulin sensitivity. As a nuclear receptor governing numerous target genes, its specific signaling transduction relies on elegant transcriptional and post-translational regulations. Notably, in response to different metabolic stimuli, PPARγ recruits various cofactors and forms distinct transcriptional complexes that change dynamically in components and epigenetic modification to ensure specific signal transduction. Clinically, PPARγ activation via its full agonists, thiazolidinediones, has been shown to improve insulin sensitivity and induce browning of white fat, while undesirably induce weight gain, visceral obesity and other adverse effects. Thus, deciphering the combinatorial interactions between PPARγ and its transcriptional partners and their preferential regulatory network in the processes of development, function and senescence of adipocytes would provide us the molecular basis for developing novel partial agonists that promote benefits of PPARγ signaling without detrimental side effects. In this review, we discuss the dynamic components and precise regulatory mechanisms of the PPARγ-cofactors complexes in adipocytes, as well as perspectives in treating metabolic diseases via specific PPARγ signaling.
Collapse
|
29
|
Affiliation(s)
- Alexandre A Steiner
- Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 1730 São Paulo, SP 05508-000, Brazil
| |
Collapse
|
30
|
Valencak TG, Osterrieder A, Schulz TJ. Sex matters: The effects of biological sex on adipose tissue biology and energy metabolism. Redox Biol 2017; 12:806-813. [PMID: 28441629 PMCID: PMC5406544 DOI: 10.1016/j.redox.2017.04.012] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/08/2017] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue is a complex and multi-faceted organ. It responds dynamically to internal and external stimuli, depending on the developmental stage and activity of the organism. The most common functional subunits of adipose tissue, white and brown adipocytes, regulate and respond to endocrine processes, which then determine metabolic rate as well as adipose tissue functions. While the molecular aspects of white and brown adipose biology have become clearer in the recent past, much less is known about sex-specific differences in regulation and deposition of adipose tissue, and the specific role of the so-called pink adipocytes during lactation in females. This review summarises the current understanding of adipose tissue dynamics with a focus on sex-specific differences in adipose tissue energy metabolism and endocrine functions, focussing on mammalian model organisms as well as human-derived data. In females, pink adipocytes trans-differentiate during pregnancy from subcutaneous white adipocytes and are responsible for milk-secretion in mammary glands. Overlooking biological sex variation may ultimately hamper clinical treatments of many aspects of metabolic disorders.
Collapse
Affiliation(s)
- Teresa G Valencak
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Veterinärplatz 1, A-1210 Vienna, Austria.
| | - Anne Osterrieder
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Headington, Oxford OX3 0BP, UK.
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, 114-116, Arthur-Scheunert-Allee, 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany.
| |
Collapse
|
31
|
Shimada IS, Acar M, Burgess RJ, Zhao Z, Morrison SJ. Prdm16 is required for the maintenance of neural stem cells in the postnatal forebrain and their differentiation into ependymal cells. Genes Dev 2017; 31:1134-1146. [PMID: 28698301 PMCID: PMC5538436 DOI: 10.1101/gad.291773.116] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 06/12/2017] [Indexed: 11/24/2022]
Abstract
Shimada et al. demonstrate that Prdm16 is required for neural stem cell maintenance and neurogenesis in the adult lateral ventricle subventricular zone and dentate gyrus. Prdm16 is also required for the formation of ciliated ependymal cells in the lateral ventricle. We and others showed previously that PR domain-containing 16 (Prdm16) is a transcriptional regulator required for stem cell function in multiple fetal and neonatal tissues, including the nervous system. However, Prdm16 germline knockout mice died neonatally, preventing us from testing whether Prdm16 is also required for adult stem cell function. Here we demonstrate that Prdm16 is required for neural stem cell maintenance and neurogenesis in the adult lateral ventricle subventricular zone and dentate gyrus. We also discovered that Prdm16 is required for the formation of ciliated ependymal cells in the lateral ventricle. Conditional Prdm16 deletion during fetal development using Nestin-Cre prevented the formation of ependymal cells, disrupting cerebrospinal fluid flow and causing hydrocephalus. Postnatal Prdm16 deletion using Nestin-CreERT2 did not cause hydrocephalus or prevent the formation of ciliated ependymal cells but caused defects in their differentiation. Prdm16 was required in neural stem/progenitor cells for the expression of Foxj1, a transcription factor that promotes ependymal cell differentiation. These studies show that Prdm16 is required for adult neural stem cell maintenance and neurogenesis as well as the formation of ependymal cells.
Collapse
Affiliation(s)
- Issei S Shimada
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Melih Acar
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Bahcesehir University, School of Medicine, Istanbul 34734, Turkey
| | - Rebecca J Burgess
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Zhiyu Zhao
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Sean J Morrison
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
32
|
Kissig M, Ishibashi J, Harms MJ, Lim HW, Stine RR, Won KJ, Seale P. PRDM16 represses the type I interferon response in adipocytes to promote mitochondrial and thermogenic programing. EMBO J 2017; 36:1528-1542. [PMID: 28408438 DOI: 10.15252/embj.201695588] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 03/15/2017] [Accepted: 03/19/2017] [Indexed: 12/13/2022] Open
Abstract
Brown adipose has the potential to counteract obesity, and thus, identifying signaling pathways that regulate the activity of this tissue is of great clinical interest. PRDM16 is a transcription factor that activates brown fat-specific genes while repressing white fat and muscle-specific genes in adipocytes. Whether PRDM16 also controls other gene programs to regulate adipocyte function was unclear. Here, we identify a novel role for PRDM16 in suppressing type I interferon (IFN)-stimulated genes (ISGs), including Stat1, in adipocytes in vitro and in vivo Ectopic activation of type I IFN signaling in brown adipocytes induces mitochondrial dysfunction and reduces uncoupling protein 1 (UCP1) expression. Prdm16-deficient adipose displays an exaggerated response to type I IFN, including higher STAT1 levels and reduced mitochondrial gene expression. Mechanistically, PRDM16 represses ISGs through binding to promoter regions of these genes and blocking the activating function of IFN regulatory factor 1 (IRF1). Together, these data indicate that PRDM16 diminishes responsiveness to type I IFN in adipose cells to promote thermogenic and mitochondrial function.
Collapse
Affiliation(s)
- Megan Kissig
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeff Ishibashi
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew J Harms
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hee-Woong Lim
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Genetics Department, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel R Stine
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kyoung-Jae Won
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Genetics Department, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA .,Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
33
|
Graus-Nunes F, Rachid TL, de Oliveira Santos F, Barbosa-da-Silva S, Souza-Mello V. AT1 receptor antagonist induces thermogenic beige adipocytes in the inguinal white adipose tissue of obese mice. Endocrine 2017; 55:786-798. [PMID: 28012150 DOI: 10.1007/s12020-016-1213-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/15/2016] [Indexed: 01/06/2023]
Abstract
PURPOSE To evaluate whether losartan is able to induce beige adipocytes formation, focusing on the thermogenic gene expression and adipocyte remodeling in the subcutaneous white adipose tissue of diet-induced obese mice. METHODS Male C57BL/6 mice received a control diet (10% energy as lipids) or a high-fat diet (50% energy as lipids) for 10 weeks, followed by a 5-week treatment with losartan: control group, control-losartan group (10 mg/Kg/day), high-fat group and high-fat-losartan group (10 mg/Kg/day). Biochemical, morphometrical, stereological and molecular approaches were used to evaluate the outcomes. RESULTS The high-fat diet elicited overweight, insulin resistance and adipocyte hypertrophy in the high-fat group, all of which losartan rescued in the high-fat-losartan group. These effects comply with the induction of beige adipocytes within the inguinal fat pads in high-fat-losartan group as they exhibited the greatest energy expenditure among the groups along with the presence uncoupling protein 1 positive multilocular adipocytes with enhanced peroxisome proliferator-activated receptor gamma coactivator 1-alpha and PR domain containing 16 mRNA levels, indicating a significant potential for mitochondrial biogenesis and adaptive thermogenesis. CONCLUSIONS Our results show compelling evidence that losartan countered diet-induced obesity in mice by enhancing energy expenditure through beige adipocytes induction. Reduced body mass, increased insulin sensitivity, decreased adipocyte size and marked expression of uncoupling protein 1 by ectopic multilocular adipocytes support these findings. The use of losartan as a coadjutant medicine to tackle obesity and its related disorders merits further investigation.
Collapse
Affiliation(s)
- Francielle Graus-Nunes
- Laboratory of Morphometry, Metabolism and Cardiovascular disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tamiris Lima Rachid
- Laboratory of Morphometry, Metabolism and Cardiovascular disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe de Oliveira Santos
- Laboratory of Morphometry, Metabolism and Cardiovascular disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sandra Barbosa-da-Silva
- Laboratory of Morphometry, Metabolism and Cardiovascular disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
34
|
Horowitz M, Kenny GP, McAllen RM, van Marken Lichtenbelt WD. Thermal physiology in a changing thermal world. Temperature (Austin) 2015; 2:22-6. [PMID: 27226998 PMCID: PMC4843882 DOI: 10.1080/23328940.2015.1017088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 02/04/2015] [Accepted: 02/04/2015] [Indexed: 12/04/2022] Open
Abstract
This editorial focuses on articles submitted to the Temperature call "Thermal Physiology in a Changing Thermal World." It highlights an array of topics related to thermoregulatory and metabolic functions in adverse environments, and the complexity and adaptability of the systems to changing climatic conditions, at various levels of body organization.
Collapse
Affiliation(s)
- Michal Horowitz
- Laboratory of Environmental Physiology; Faculty of Dentistry; The Hebrew University of Jerusalem; Israel
| | - Glen P Kenny
- Human Environmental Physiological Research Unit; University of Ottawa; Canada
| | - Robin M McAllen
- The Florey Institute of Neuroscience and Mental Health; University of Melbourne; Melbourne, VIC Australia
- Department of Anatomy & Neuroscience; University Of Melbourne; Melbourne, VIC Australia
| | - Wouter D van Marken Lichtenbelt
- Department of Human Biology; NUTRIM School for Nutrition, Toxicology and Metabolism of Maastricht University Medical Center; The Netherlands
| |
Collapse
|