1
|
Sabatino L, Vassalle C. Thyroid Hormones and Metabolism Regulation: Which Role on Brown Adipose Tissue and Browning Process? Biomolecules 2025; 15:361. [PMID: 40149897 PMCID: PMC11940499 DOI: 10.3390/biom15030361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Thyroid hormones (THs) are important modulators of many metabolic processes, being strictly associated with the control of energy balance, mainly through activities on the brain, white and brown adipose tissue, skeletal muscle, liver, and pancreas. In this review, the principal mechanisms of TH regulation on metabolic processes will be discussed and THs' relevance in metabolic disease progression will be evaluated, especially in the cardiovascular context and correlated diseases. Moreover, we will discuss THs' regulatory role on metabolic events in white and brown adipose tissue, with a special focus on the process of "browning", which consists of the gradual acquisition by white adipocytes of the physical and functional characteristics of brown adipocytes. The advancements in research on molecular mechanisms and proposed physiopathological relevance of this process will be discussed.
Collapse
Affiliation(s)
- Laura Sabatino
- Institute of Clinical Physiology, National Council of Research, 56124 Pisa, Italy
| | | |
Collapse
|
2
|
Roth L, Hoffmann A, Hagemann T, Wagner L, Strehlau C, Sheikh B, Donndorf L, Ghosh A, Noé F, Wolfrum C, Krohn K, Weiner J, Heiker JT, Klöting N, Stumvoll M, Tönjes A, Blüher M, Mittag J, Krause K. Thyroid hormones are required for thermogenesis of beige adipocytes induced by Zfp423 inactivation. Cell Rep 2024; 43:114987. [PMID: 39580797 DOI: 10.1016/j.celrep.2024.114987] [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: 03/20/2024] [Revised: 08/30/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024] Open
Abstract
The significance of thyroid hormones (THs) in beige adipocyte thermogenesis remains incompletely understood. We previously reported that THs directly regulate the expression of zinc-finger protein 423 (ZFP423), an anti-thermogenic factor, in adipose tissue. This study investigates the interaction between THs and adrenergic signaling in regulating thermogenic capacity and activation of beige adipocytes formed in response to Zfp423 deletion. We demonstrate that THs are indispensable for uncoupling protein 1 (UCP1)-dependent thermogenesis, leading to increased energy expenditure in mice with adipocyte-specific Zfp423 knockout. Targeted activation of the thyroid receptor isoform TRβ, which plays a central role in the inguinal depot, is sufficient to enhance energy expenditure in hypothyroid Zfp423iAKO mice. Mechanistically, THs and ZFP423 pathways cooperate to regulate early B cell factor 2 (EBF2)-mediated activation of the Ucp1 gene. RNA sequencing (RNA-seq) analysis of human adipose tissue samples supports the relevance of this regulatory network for human adipose tissue plasticity.
Collapse
Affiliation(s)
- Lisa Roth
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Tobias Hagemann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Leonie Wagner
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Christian Strehlau
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Bilal Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Lorenz Donndorf
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Adhideb Ghosh
- Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zürich, Switzerland
| | - Falko Noé
- Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zürich, Switzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zürich, Switzerland
| | - Knut Krohn
- Medical Faculty, Center for DNA Technologies, University of Leipzig, 04103 Leipzig, Germany
| | - Juliane Weiner
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - John T Heiker
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Michael Stumvoll
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Anke Tönjes
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Matthias Blüher
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Jens Mittag
- Institute of Experimental Endocrinology/CBBM, University of Lübeck, 23562 Lübeck, Germany
| | - Kerstin Krause
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; German Center for Diabetes Research e.V., 85764 Neuherberg, Germany.
| |
Collapse
|
3
|
Huang L, Guo Z, Huang M, Zeng X, Huang H. Triiodothyronine (T3) promotes browning of white adipose through inhibition of the PI3K/AKT signalling pathway. Sci Rep 2024; 14:20370. [PMID: 39223267 PMCID: PMC11369215 DOI: 10.1038/s41598-024-71591-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024] Open
Abstract
Obesity arises from an imbalance between energy consumption and energy expenditure, and thyroid hormone levels serve as a determinant of energy expenditure. We conducted experiments at the animal and cellular levels and combined those findings with clinical data to elucidate the role of triiodothyronine (T3) in facilitating the browning of white adipose tissue (WAT) and its underlying mechanism. The results showed (i) the impaired metabolic function of local WAT and the compensatory elevation of systemic thermogenesis in obesity; (ii) T3 treatment of white adipocytes in vitro and local WAT in vivo induced a shift towards a morphologically "brown" phenotype, accompanied by upregulation of mRNA and protein expression of browning-related and mitochondrial function markers, which suggest that T3 intervention promotes the browning of WAT; and (iii) the aforementioned processes could be modulated through inhibition of the PI3K/AKT signalling pathway; however, whether T3 affects the PI3K/AKT signalling pathway by affecting insulin signalling remains to be studied and clarified. The results of our study indicate that T3 treatment promotes browning of WAT through inhibition of the PI3K/AKT signalling pathway; these findings offer novel perspectives regarding the potential of localised therapies for addressing WAT volume in individuals with obesity.
Collapse
Affiliation(s)
- LingHong Huang
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - ZhiFeng Guo
- Department of Respiratory Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - MingJing Huang
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - XiYing Zeng
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - HuiBin Huang
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China.
| |
Collapse
|
4
|
Emamnejad R, Pagnin M, Petratos S. The iron maiden: Oligodendroglial metabolic dysfunction in multiple sclerosis and mitochondrial signaling. Neurosci Biobehav Rev 2024; 164:105788. [PMID: 38950685 DOI: 10.1016/j.neubiorev.2024.105788] [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: 04/02/2024] [Revised: 06/06/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune disease, governed by oligodendrocyte (OL) dystrophy and central nervous system (CNS) demyelination manifesting variable neurological impairments. Mitochondrial mechanisms may drive myelin biogenesis maintaining the axo-glial unit according to dynamic requisite demands imposed by the axons they ensheath. The promotion of OL maturation and myelination by actively transporting thyroid hormone (TH) into the CNS and thereby facilitating key transcriptional and metabolic pathways that regulate myelin biogenesis is fundamental to sustain the profound energy demands at each axo-glial interface. Deficits in regulatory functions exerted through TH for these physiological roles to be orchestrated by mature OLs, can occur in genetic and acquired myelin disorders, whereby mitochondrial efficiency and eventual dysfunction can lead to profound oligodendrocytopathy, demyelination and neurodegenerative sequelae. TH-dependent transcriptional and metabolic pathways can be dysregulated during acute and chronic MS lesion activity depriving OLs from critical acetyl-CoA biochemical mechanisms governing myelin lipid biosynthesis and at the same time altering the generation of iron metabolism that may drive ferroptotic mechanisms, leading to advancing neurodegeneration.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| |
Collapse
|
5
|
Ruswandi YAR, Lesmana R, Rosdianto AM, Gunadi JW, Goenawan H, Zulhendri F. Understanding the Roles of Selenium on Thyroid Hormone-Induced Thermogenesis in Adipose Tissue. Biol Trace Elem Res 2024; 202:2419-2441. [PMID: 37758980 DOI: 10.1007/s12011-023-03854-2] [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: 04/18/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
Brown adipose tissue (BAT) and white adipose tissue (WAT) are known to regulate lipid metabolism. A lower amount of BAT compared to WAT, along with adipose tissue dysfunction, can result in obesity. Studies have shown that selenium supplementation protects against adipocyte dysfunction, decreases WAT triglycerides, and increases BAT triiodothyronine (T3). In this review, we discuss the relationship between selenium and lipid metabolism regulation through selenoprotein deiodinases and the role of deiodinases and thyroid hormones in the induction of adipose tissue thermogenesis. Upon 22 studies included in our review, we found that studies investigating the relationship between selenium and deiodinases demonstrated that selenium supplementation affects the iodothyronine deiodinase 2 (DIO2) protein and the expression of its associated gene, DIO2, proportionally. However, its effect on DIO1 is inconsistent while its effect on DIO3 activity is not detected. Studies have shown that the activity of deiodinases especially DIO2 protein and DIO2 gene expression is increased along with other browning markers upon white adipose tissue browning induction. Studies showed that thermogenesis is stimulated by the thyroid hormone T3 as its activity is correlated to the expression of other thermogenesis markers. A proposed mechanism of thermogenesis induction in selenium supplementation is by autophagy control. However, more studies are needed to establish the role of T3 and autophagy in adipose tissue thermogenesis, especially, since some studies have shown that thermogenesis can function even when T3 activity is lacking and studies related to autophagy in adipose tissue thermogenesis have contradictory results.
Collapse
Affiliation(s)
- Yasmin Anissa R Ruswandi
- Graduate School of Master Program in Anti-Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Kabupaten Sumedang, West Java, Indonesia
| | - Ronny Lesmana
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang, KM.21, Hegarmanah, Kec. Jatinangor, Kabupaten Sumedang, West Java, 45363, Indonesia.
| | - Aziiz Mardanarian Rosdianto
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang, KM.21, Hegarmanah, Kec. Jatinangor, Kabupaten Sumedang, West Java, 45363, Indonesia
- Veterinary Medicine Study Program, Faculty of Medicine, Universitas Padjadjaran, Kabupaten Sumedang, West Java, Indonesia
| | - Julia Windi Gunadi
- Department of Physiology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Hanna Goenawan
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang, KM.21, Hegarmanah, Kec. Jatinangor, Kabupaten Sumedang, West Java, 45363, Indonesia
| | - Felix Zulhendri
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Kabupaten Sumedang, West Java, Indonesia
- Kebun Efi, Kabanjahe, 22171, North Sumatra, Indonesia
| |
Collapse
|
6
|
Ghesmati Z, Rashid M, Fayezi S, Gieseler F, Alizadeh E, Darabi M. An update on the secretory functions of brown, white, and beige adipose tissue: Towards therapeutic applications. Rev Endocr Metab Disord 2024; 25:279-308. [PMID: 38051471 PMCID: PMC10942928 DOI: 10.1007/s11154-023-09850-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 12/07/2023]
Abstract
Adipose tissue, including white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue, is vital in modulating whole-body energy metabolism. While WAT primarily stores energy, BAT dissipates energy as heat for thermoregulation. Beige adipose tissue is a hybrid form of adipose tissue that shares characteristics with WAT and BAT. Dysregulation of adipose tissue metabolism is linked to various disorders, including obesity, type 2 diabetes, cardiovascular diseases, cancer, and infertility. Both brown and beige adipocytes secrete multiple molecules, such as batokines, packaged in extracellular vesicles or as soluble signaling molecules that play autocrine, paracrine, and endocrine roles. A greater understanding of the adipocyte secretome is essential for identifying novel molecular targets in treating metabolic disorders. Additionally, microRNAs show crucial roles in regulating adipose tissue differentiation and function, highlighting their potential as biomarkers for metabolic disorders. The browning of WAT has emerged as a promising therapeutic approach in treating obesity and associated metabolic disorders. Many browning agents have been identified, and nanotechnology-based drug delivery systems have been developed to enhance their efficacy. This review scrutinizes the characteristics of and differences between white, brown, and beige adipose tissues, the molecular mechanisms involved in the development of the adipocytes, the significant roles of batokines, and regulatory microRNAs active in different adipose tissues. Finally, the potential of WAT browning in treating obesity and atherosclerosis, the relationship of BAT with cancer and fertility disorders, and the crosstalk between adipose tissue with circadian system and circadian disorders are also investigated.
Collapse
Affiliation(s)
- Zeinab Ghesmati
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashid
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Fayezi
- Department of Gynecologic Endocrinology and Fertility Disorders, Women's Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Frank Gieseler
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany.
| |
Collapse
|
7
|
Sinha RA, Yen PM. Metabolic Messengers: Thyroid Hormones. Nat Metab 2024; 6:639-650. [PMID: 38671149 PMCID: PMC7615975 DOI: 10.1038/s42255-024-00986-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 01/15/2024] [Indexed: 04/28/2024]
Abstract
Thyroid hormones (THs) are key hormones that regulate development and metabolism in mammals. In man, the major target tissues for TH action are the brain, liver, muscle, heart, and adipose tissue. Defects in TH synthesis, transport, metabolism, and nuclear action have been associated with genetic and endocrine diseases in man. Over the past few years, there has been renewed interest in TH action and the therapeutic potential of THs and thyromimetics to treat several metabolic disorders such as hypercholesterolemia, dyslipidaemia, non-alcoholic fatty liver disease (NAFLD), and TH transporter defects. Recent advances in the development of tissue and TH receptor isoform-targeted thyromimetics have kindled new hope for translating our fundamental understanding of TH action into an effective therapy. This review provides a concise overview of the historical development of our understanding of TH action, its physiological and pathophysiological effects on metabolism, and future therapeutic applications to treat metabolic dysfunction.
Collapse
Affiliation(s)
- Rohit A Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
| | - Paul M Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore.
- Div. Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
8
|
Abbasi K, Zarezadeh R, Valizadeh A, Mehdizadeh A, Hamishehkar H, Nouri M, Darabi M. White-brown adipose tissue interplay in polycystic ovary syndrome: Therapeutic avenues. Biochem Pharmacol 2024; 220:116012. [PMID: 38159686 DOI: 10.1016/j.bcp.2023.116012] [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: 11/28/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
This study highlights the therapeutic potential of activating brown adipose tissue (BAT) for managing polycystic ovary syndrome (PCOS), a prevalent endocrine disorder associated with metabolic and reproductive abnormalities. BAT plays a crucial role in regulating energy expenditure and systemic insulin sensitivity, making it an attractive target for the treatment of obesity and metabolic diseases. Recent research suggests that impaired BAT function and mass may contribute to the link between metabolic disturbances and reproductive issues in PCOS. Additionally, abnormal white adipose tissue (WAT) can exacerbate these conditions by releasing adipokines and nonesterified fatty acids. In this review, we explored the impact of WAT changes on BAT function in PCOS and discussed the potential of BAT activation as a therapeutic strategy to improve PCOS symptoms. We propose that BAT activation holds promise for managing PCOS; however, further research is needed to confirm its efficacy and to develop clinically feasible methods for BAT activation.
Collapse
Affiliation(s)
- Khadijeh Abbasi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Valizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, Germany.
| |
Collapse
|
9
|
Mosteiro L, Nguyen TTT, Hankeova S, Alvarez-Sierra D, Reichelt M, Vandriel SM, Lai Z, Choudhury FK, Sangaraju D, Kamath BM, Scherl A, Pujol-Borrell R, Piskol R, Siebel CW. Notch signaling in thyrocytes is essential for adult thyroid function and mammalian homeostasis. Nat Metab 2023; 5:2094-2110. [PMID: 38123718 DOI: 10.1038/s42255-023-00937-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/31/2023] [Indexed: 12/23/2023]
Abstract
The thyroid functions as an apex endocrine organ that controls growth, differentiation and metabolism1, and thyroid diseases comprise the most common endocrine disorders2. Nevertheless, high-resolution views of the cellular composition and signals that govern the thyroid have been lacking3,4. Here, we show that Notch signalling controls homeostasis and thermoregulation in adult mammals through a mitochondria-based mechanism in a subset of thyrocytes. We discover two thyrocyte subtypes in mouse and human thyroids, identified in single-cell analyses by different levels of metabolic activity and Notch signalling. Therapeutic antibody blockade of Notch in adult mice inhibits a thyrocyte-specific transcriptional program and induces thyrocyte defects due to decreased mitochondrial activity and ROS production. Thus, disrupting Notch signalling in adult mice causes hypothyroidism, characterized by reduced levels of circulating thyroid hormone and dysregulation of whole-body thermoregulation. Inducible genetic deletion of Notch1 and 2 in thyrocytes phenocopies this antibody-induced hypothyroidism, establishing a direct role for Notch in adult murine thyrocytes. We confirm that hypothyroidism is enriched in children with Alagille syndrome, a genetic disorder marked by Notch mutations, suggesting that these findings translate to humans.
Collapse
Grants
- NA Genentech (Genentech, Inc.)
- NA Genentech (Genentech, Inc.)
- NA Genentech (Genentech, Inc.)
- NA Genentech (Genentech, Inc.)
- NA Genentech (Genentech, Inc.)
- NA Genentech (Genentech, Inc.)
- NA Genentech (Genentech, Inc.)
- NA Genentech (Genentech, Inc.)
- NA Genentech (Genentech, Inc.)
- NA Genentech (Genentech, Inc.)
Collapse
Affiliation(s)
- Lluc Mosteiro
- Department of Discovery Oncology, Genentech, South San Francisco, CA, USA.
| | - Thi Thu Thao Nguyen
- Department of Oncology Bioinformatics, Genentech, South San Francisco, CA, USA
| | - Simona Hankeova
- Department of Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Daniel Alvarez-Sierra
- Translational Immunology Group, Vall d'Hebron Institut de Recerca (VHIR), Campus Vall Hebron, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Mike Reichelt
- Department of Research Pathology, Genentech, South San Francisco, CA, USA
| | - Shannon M Vandriel
- Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Zijuan Lai
- Department of Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, CA, USA
| | - Feroza K Choudhury
- Department of Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, CA, USA
| | - Dewakar Sangaraju
- Department of Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, CA, USA
| | - Binita M Kamath
- Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Alexis Scherl
- Department of Research Pathology, Genentech, South San Francisco, CA, USA
| | - Ricardo Pujol-Borrell
- Translational Immunology Group, Vall d'Hebron Institut de Recerca (VHIR), Campus Vall Hebron, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Vall Hebron Institute of Oncology (VHIO), Campus Vall Hebron, Barcelona, Spain
| | - Robert Piskol
- Department of Oncology Bioinformatics, Genentech, South San Francisco, CA, USA
| | - Christian W Siebel
- Department of Discovery Oncology, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
10
|
Lundgren P, Sharma PV, Dohnalová L, Coleman K, Uhr GT, Kircher S, Litichevskiy L, Bahnsen K, Descamps HC, Demetriadou C, Chan J, Chellappa K, Cox TO, Heyman Y, Pather SR, Shoffler C, Petucci C, Shalem O, Raj A, Baur JA, Snyder NW, Wellen KE, Levy M, Seale P, Li M, Thaiss CA. A subpopulation of lipogenic brown adipocytes drives thermogenic memory. Nat Metab 2023; 5:1691-1705. [PMID: 37783943 PMCID: PMC11309804 DOI: 10.1038/s42255-023-00893-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/21/2023] [Indexed: 10/04/2023]
Abstract
Sustained responses to transient environmental stimuli are important for survival. The mechanisms underlying long-term adaptations to temporary shifts in abiotic factors remain incompletely understood. Here, we find that transient cold exposure leads to sustained transcriptional and metabolic adaptations in brown adipose tissue, which improve thermogenic responses to secondary cold encounter. Primary thermogenic challenge triggers the delayed induction of a lipid biosynthesis programme even after cessation of the original stimulus, which protects from subsequent exposures. Single-nucleus RNA sequencing and spatial transcriptomics reveal that this response is driven by a lipogenic subpopulation of brown adipocytes localized along the perimeter of Ucp1hi adipocytes. This lipogenic programme is associated with the production of acylcarnitines, and supplementation of acylcarnitines is sufficient to recapitulate improved secondary cold responses. Overall, our data highlight the importance of heterogenous brown adipocyte populations for 'thermogenic memory', which may have therapeutic implications for leveraging short-term thermogenesis to counteract obesity.
Collapse
Affiliation(s)
- Patrick Lundgren
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Prateek V Sharma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Lenka Dohnalová
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kyle Coleman
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Giulia T Uhr
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susanna Kircher
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lev Litichevskiy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Klaas Bahnsen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hélène C Descamps
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christina Demetriadou
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jacqueline Chan
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karthikeyani Chellappa
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Timothy O Cox
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yael Heyman
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarshan R Pather
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Clarissa Shoffler
- Penn Metabolomics Core, Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher Petucci
- Penn Metabolomics Core, Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Ophir Shalem
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Arjun Raj
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Baur
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathaniel W Snyder
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Maayan Levy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Seale
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Development Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Xue S, Lee D, Berry DC. Thermogenic adipose tissue in energy regulation and metabolic health. Front Endocrinol (Lausanne) 2023; 14:1150059. [PMID: 37020585 PMCID: PMC10067564 DOI: 10.3389/fendo.2023.1150059] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
The ability to generate thermogenic fat could be a targeted therapy to thwart obesity and improve metabolic health. Brown and beige adipocytes are two types of thermogenic fat cells that regulate energy balance. Both adipocytes share common morphological, biochemical, and thermogenic properties. Yet, recent evidence suggests unique features exist between brown and beige adipocytes, such as their cellular origin and thermogenic regulatory processes. Beige adipocytes also appear highly plastic, responding to environmental stimuli and interconverting between beige and white adipocyte states. Additionally, beige adipocytes appear to be metabolically heterogenic and have substrate specificity. Nevertheless, obese and aged individuals cannot develop beige adipocytes in response to thermogenic fat-inducers, creating a key clinical hurdle to their therapeutic promise. Thus, elucidating the underlying developmental, molecular, and functional mechanisms that govern thermogenic fat cells will improve our understanding of systemic energy regulation and strive for new targeted therapies to generate thermogenic fat. This review will examine the recent advances in thermogenic fat biogenesis, molecular regulation, and the potential mechanisms for their failure.
Collapse
Affiliation(s)
| | | | - Daniel C. Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
12
|
Roth L, Johann K, Hönes GS, Oelkrug R, Wagner L, Hoffmann A, Krohn K, Moeller LC, Weiner J, Heiker JT, Klöting N, Tönjes A, Stumvoll M, Blüher M, Mittag J, Krause K. Thyroid hormones regulate Zfp423 expression in regionally distinct adipose depots through direct and cell-autonomous action. Cell Rep 2023; 42:112088. [PMID: 36753417 DOI: 10.1016/j.celrep.2023.112088] [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: 03/23/2022] [Revised: 12/05/2022] [Accepted: 01/24/2023] [Indexed: 02/09/2023] Open
Abstract
The hypothalamic pituitary thyroid axis is a major regulator of many differentiation processes, including adipose tissue. However, it remains unclear whether and how thyroid hormone (TH) signaling contributes to preadipocyte commitment and differentiation into mature adipocytes. Here, we show a cell-autonomous effect of TH on the transcriptional regulation of zinc finger protein 423 (Zfp423), an early adipogenic determination factor, in murine adipose depots. Mechanistically, binding of the unliganded TH receptor to a negative TH responsive element within the Zfp423 promoter activates transcriptional activity that is reversed upon TH binding. Zfp423 upregulation is associated with increased GFP+ preadipocyte recruitment in stromal vascular fraction isolated from white fat of hypothyroid Zfp423GFP reporter mice. RNA sequencing identified Zfp423-driven gene programs that are modulated in response to TH during adipogenic differentiation. Collectively, we identified Zfp423 as a key molecule that integrates TH signaling into the regulation of adipose tissue plasticity.
Collapse
Affiliation(s)
- Lisa Roth
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Kornelia Johann
- Institute for Endocrinology and Diabetes/Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Georg Sebastian Hönes
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Rebecca Oelkrug
- Institute for Endocrinology and Diabetes/Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Leonie Wagner
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Knut Krohn
- DNA Core Unit Leipzig, University of Leipzig, 04103 Leipzig, Germany
| | - Lars C Moeller
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Juliane Weiner
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - John T Heiker
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Anke Tönjes
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Michael Stumvoll
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Matthias Blüher
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Jens Mittag
- Institute for Endocrinology and Diabetes/Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Kerstin Krause
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; Deutsches Zentrum für Diabetesforschung e.V., 85764 Neuherberg, Germany.
| |
Collapse
|
13
|
Recinella L, De Filippis B, Libero ML, Ammazzalorso A, Chiavaroli A, Orlando G, Ferrante C, Giampietro L, Veschi S, Cama A, Mannino F, Gasparo I, Bitto A, Amoroso R, Brunetti L, Leone S. Anti-Inflammatory, Antioxidant, and WAT/BAT-Conversion Stimulation Induced by Novel PPAR Ligands: Results from Ex Vivo and In Vitro Studies. Pharmaceuticals (Basel) 2023; 16:346. [PMID: 36986448 PMCID: PMC10056895 DOI: 10.3390/ph16030346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Activation of peroxisome proliferator-activated receptors (PPARs) not only regulates multiple metabolic pathways, but mediates various biological effects related to inflammation and oxidative stress. We investigated the effects of four new PPAR ligands containing a fibrate scaffold-the PPAR agonists (1a (αEC50 1.0 μM) and 1b (γEC50 0.012 μM)) and antagonists (2a (αIC50 6.5 μM) and 2b (αIC50 0.98 μM, with a weak antagonist activity on γ isoform))-on proinflammatory and oxidative stress biomarkers. The PPAR ligands 1a-b and 2a-b (0.1-10 μM) were tested on isolated liver specimens treated with lipopolysaccharide (LPS), and the levels of lactate dehydrogenase (LDH), prostaglandin (PG) E2, and 8-iso-PGF2α were measured. The effects of these compounds on the gene expression of the adipose tissue markers of browning, PPARα, and PPARγ, in white adipocytes, were evaluated as well. We found a significant reduction in LPS-induced LDH, PGE2, and 8-iso-PGF2α levels after 1a treatment. On the other hand, 1b decreased LPS-induced LDH activity. Compared to the control, 1a stimulated uncoupling protein 1 (UCP1), PR-(PRD1-BF1-RIZ1 homologous) domain containing 16 (PRDM16), deiodinase type II (DIO2), and PPARα and PPARγ gene expression, in 3T3-L1 cells. Similarly, 1b increased UCP1, DIO2, and PPARγ gene expression. 2a-b caused a reduction in the gene expression of UCP1, PRDM16, and DIO2 when tested at 10 μM. In addition, 2a-b significantly decreased PPARα gene expression. A significant reduction in PPARγ gene expression was also found after 2b treatment. The novel PPARα agonist 1a might be a promising lead compound and represents a valuable pharmacological tool for further assessment. The PPARγ agonist 1b could play a minor role in the regulation of inflammatory pathways.
Collapse
Affiliation(s)
- Lucia Recinella
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | | | | | | | | | - Giustino Orlando
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | - Claudio Ferrante
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | | | - Serena Veschi
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | - Alessandro Cama
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | - Federica Mannino
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Irene Gasparo
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Rosa Amoroso
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | - Luigi Brunetti
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | - Sheila Leone
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| |
Collapse
|
14
|
COBL, MKX and MYOC Are Potential Regulators of Brown Adipose Tissue Development Associated with Obesity-Related Metabolic Dysfunction in Children. Int J Mol Sci 2023; 24:ijms24043085. [PMID: 36834493 PMCID: PMC9964948 DOI: 10.3390/ijms24043085] [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: 12/23/2022] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Obesity is already accompanied by adipose tissue (AT) dysfunction and metabolic disease in children and increases the risk of premature death. Due to its energy-dissipating function, brown AT (BAT) has been discussed as being protective against obesity and related metabolic dysfunction. To analyze the molecular processes associated with BAT development, we investigated genome-wide expression profiles in brown and white subcutaneous and perirenal AT samples of children. We identified 39 upregulated and 26 downregulated genes in uncoupling protein 1 (UCP1)-positive compared to UCP1-negative AT samples. We prioritized for genes that had not been characterized regarding a role in BAT biology before and selected cordon-bleu WH2 repeat protein (COBL), mohawk homeobox (MKX) and myocilin (MYOC) for further functional characterization. The siRNA-mediated knockdown of Cobl and Mkx during brown adipocyte differentiation in vitro resulted in decreased Ucp1 expression, while the inhibition of Myoc led to increased Ucp1 expression. Furthermore, COBL, MKX and MYOC expression in the subcutaneous AT of children is related to obesity and parameters of AT dysfunction and metabolic disease, such as adipocyte size, leptin levels and HOMA-IR. In conclusion, we identify COBL, MKX and MYOC as potential regulators of BAT development and show an association of these genes with early metabolic dysfunction in children.
Collapse
|
15
|
Wen X, Ding X, Chang X, Wang J, Wang Q, Liu J, Wang G. Elevated serum Meteorin-like levels in patients with hyperthyroidism. BMC Endocr Disord 2022; 22:304. [PMID: 36476346 PMCID: PMC9727892 DOI: 10.1186/s12902-022-01229-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Meteorin-like (Metrnl) is a newly discovered adipomyokine that regulates systemic energy homeostasis. Both thyroid hormones and Metrnl increase energy expenditure and induce browning of adipose tissue. Thus, the aim of this study was to investigate serum Metrnl levels in hyperthyroid patients and the association of serum Metrnl levels with hyperthyroidism. METHODS The study included 88 patients with newly diagnosed untreated overt hyperthyroidism and 100 age- and sex- matched healthy controls. Serum Metrnl levels were determined using the enzyme-linked immunosorbent assay (ELISA) method. RESULTS Serum Metrnl levels were significantly elevated in patients with hyperthyroidism compared with controls. Linear regression analyses indicated that serum Metrnl levels were independently associated with FT3 (β = 0.324, P = 0.001), FT4 (β = 0.293, P = 0.001), and TSH (β = -0.234, P = 0.006) after full adjustment. Additionally, further logistic regression analyses revealed that the highest Metrnl tertile was significantly associated with hyperthyroidism compared with the lowest tertile (P for trend < 0.001). The relationship remained significant even after adjusting for potential confounders. Meanwhile, each one-unit increase in circulating Metrnl was independently associated with hyperthyroidism (OR 1.021, 95%CI 1.007-1.036, P < 0.01). CONCLUSION Serum Metrnl levels were elevated in patients with hyperthyroidism and were independently associated with hyperthyroidism.
Collapse
Affiliation(s)
- Xiaohui Wen
- Department of Otolaryngology Head & Neck Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xiaoyu Ding
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Xiaona Chang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Jiaxuan Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Qiu Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China.
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, NO. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
16
|
Ho WE, Sun L, Goh HJ, Tint MT, Sun L, Leow MKS. Brown adipose tissue influences adiponectin and thyroid hormone changes during Graves' disease therapy. Adipocyte 2022; 11:389-400. [PMID: 35894647 PMCID: PMC9336474 DOI: 10.1080/21623945.2022.2104509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Thyroid hormones (TH), adiponectin and brown adipose tissue (BAT) are regulators of energy homoeostasis. Influence of BAT activity on the relationship between TH and adiponectin remains unexplored. The aim of the study was to identify the relationship between TH and adiponectin and to clarify the impact of active BAT on the metabolic effects of adiponectin before and after the correction of thyrotoxicosis. Twenty-one patients with newly diagnosed hyperthyroidism from Graves' disease were recruited. A titration dosing regimen of thionamide anti-thyroid drug (ATD) was used to establish euthyroidism over 12-24 weeks. Anthropometric, biochemical and adipocytokine parameters were measured before and after control of hyperthyroidism. BAT activity was quantified by fusion 18 F-fluorodeoxyglucose (18 F-FDG) PET/MR imaging, and patients were grouped based on BAT status. Plasma adiponectin level was significantly increased following correction of hyperthyroidism in the overall sample. Free thyroxine (FT4) was also identified as a predictor of adiponectin level in thyroid dysfunction. However, significant changes in adiponectin level and correlations involving adiponectin were absent in BAT-positive patients but maintained in BAT-negative patients. BAT activity diminishes the correlative relationship with body composition and abolishes TH and adiponectin relationships when transitioning from a hyperthyroid to euthyroid state.
Collapse
Affiliation(s)
- Wei-En Ho
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore, Singapore
| | - Lijuan Sun
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A *STAR), Singapore
| | - Hui Jen Goh
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A *STAR), Singapore
| | - Mya Thway Tint
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A *STAR), Singapore.,Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Melvin Khee Shing Leow
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore, Singapore.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A *STAR), Singapore.,Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Endocrinology, Division of Medicine, Tan Tock Seng Hospital (TTSH), Singapore
| |
Collapse
|
17
|
Christen L, Broghammer H, Rapöhn I, Möhlis K, Strehlau C, Ribas‐Latre A, Gebhardt C, Roth L, Krause K, Landgraf K, Körner A, Rohde‐Zimmermann K, Hoffmann A, Klöting N, Ghosh A, Sun W, Dong H, Wolfrum C, Rassaf T, Hendgen‐Cotta UB, Stumvoll M, Blüher M, Heiker JT, Weiner J. Myoglobin-mediated lipid shuttling increases adrenergic activation of brown and white adipocyte metabolism and is as a marker of thermogenic adipocytes in humans. Clin Transl Med 2022; 12:e1108. [PMID: 36480426 PMCID: PMC9731393 DOI: 10.1002/ctm2.1108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recruitment and activation of brown adipose tissue (BAT) results in increased energy expenditure (EE) via thermogenesis and represents an intriguing therapeutic approach to combat obesity and treat associated diseases. Thermogenesis requires an increased and efficient supply of energy substrates and oxygen to the BAT. The hemoprotein myoglobin (MB) is primarily expressed in heart and skeletal muscle fibres, where it facilitates oxygen storage and flux to the mitochondria during exercise. In the last years, further contributions of MB have been assigned to the scavenging of reactive oxygen species (ROS), the regulation of cellular nitric oxide (NO) levels and also lipid binding. There is a substantial expression of MB in BAT, which is induced during brown adipocyte differentiation and BAT activation. This suggests MB as a previously unrecognized player in BAT contributing to thermogenesis. METHODS AND RESULTS This study analyzed the consequences of MB expression in BAT on mitochondrial function and thermogenesis in vitro and in vivo. Using MB overexpressing, knockdown or knockout adipocytes, we show that expression levels of MB control brown adipocyte mitochondrial respiratory capacity and acute response to adrenergic stimulation, signalling and lipolysis. Overexpression in white adipocytes also increases their metabolic activity. Mutation of lipid interacting residues in MB abolished these beneficial effects of MB. In vivo, whole-body MB knockout resulted in impaired thermoregulation and cold- as well as drug-induced BAT activation in mice. In humans, MB is differentially expressed in subcutaneous (SC) and visceral (VIS) adipose tissue (AT) depots, differentially regulated by the state of obesity and higher expressed in AT samples that exhibit higher thermogenic potential. CONCLUSIONS These data demonstrate for the first time a functional relevance of MBs lipid binding properties and establish MB as an important regulatory element of thermogenic capacity in brown and likely beige adipocytes.
Collapse
Affiliation(s)
- Lisa Christen
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Helen Broghammer
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Inka Rapöhn
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| | - Kevin Möhlis
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Christian Strehlau
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| | - Aleix Ribas‐Latre
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Claudia Gebhardt
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Lisa Roth
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| | - Kerstin Krause
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| | - Kathrin Landgraf
- Center for Pediatric Research Leipzig (CPL)University Hospital for Children and AdolescentsMedical FacultyUniversity of LeipzigLeipzigGermany
| | - Antje Körner
- Center for Pediatric Research Leipzig (CPL)University Hospital for Children and AdolescentsMedical FacultyUniversity of LeipzigLeipzigGermany
| | - Kerstin Rohde‐Zimmermann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Adhideb Ghosh
- Institute of FoodNutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Wenfei Sun
- Institute of FoodNutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Hua Dong
- Institute of FoodNutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Christian Wolfrum
- Institute of FoodNutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Tienush Rassaf
- Department of Cardiology and Vascular MedicineWest German Heart and Vascular CenterMedical FacultyUniversity of Duisburg‐EssenEssenGermany
| | - Ulrike B. Hendgen‐Cotta
- Department of Cardiology and Vascular MedicineWest German Heart and Vascular CenterMedical FacultyUniversity of Duisburg‐EssenEssenGermany
| | - Michael Stumvoll
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - John T. Heiker
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
- Institute of Biochemistry, Faculty of Life SciencesUniversity of LeipzigLeipzigGermany
| | - Juliane Weiner
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| |
Collapse
|
18
|
Choi J, Lee K, Kim J, Jeong W, Jo T, Lee HW, Park YS, Park SW. Thyroid Hormone Ameliorates Lymphedema by Suppressing Adipogenesis in a Murine Lymphedema Model. Lymphat Res Biol 2022; 20:585-592. [PMID: 35333603 DOI: 10.1089/lrb.2021.0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background: Exogenous supplementation of thyroid hormone could inhibit excessive fat deposition in lymphedema tissue by suppressing adipogenesis. Methods and Results: Cell viability, adipogenic differentiation, and mRNA expression were measured in 3T3-L1 preadipocytes treated with L-thyroxine. Twelve mice were divided into control and L-thyroxine groups. Two weeks after lymphedema was surgically induced, the experimental mice were fed L-thyroxine for 4 weeks. Tail volume and body weight were measured, and 6 weeks after the surgery, tail skin and subcutaneous tissue were harvested for histopathologic examination and protein isolation. In 3T3-L1 cells, treatment with 10-500 μM L-thyroxine did not affect cell viability. Eight days after induction of adipogenic differentiation, lipid accumulation decreased significantly in the 50 and 100 μM L-thyroxine groups (p < 0.001). mRNA levels of peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer binding protein α (C/EBPα), and fatty acid-binding protein 4 (FABP4) decreased significantly in the 100 μM L-thyroxine group compared with the control group (p = 0.017). Lymphedema tails treated with L-thyroxine exhibited decreased volume (p = 0.028) and thickness of dermal and subcutaneous tissue (p = 0.01) and increased vascular endothelial growth factor-C protein expression (p = 0.017) compared with the control. Conclusion: Thyroid hormone therapy inhibits the adipogenesis of 3T3-L1 cells in vitro and decreases the volume of murine lymphedema tail in vivo. These findings suggest that thyroid hormone therapy could be used to treat lymphedema.
Collapse
Affiliation(s)
- Jaehoon Choi
- Department of Plastic and Reconstructive Surgery, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Kanghee Lee
- Department of Plastic and Reconstructive Surgery, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Junhyung Kim
- Department of Plastic and Reconstructive Surgery, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Woonhyeok Jeong
- Department of Plastic and Reconstructive Surgery, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Taehee Jo
- Department of Plastic and Reconstructive Surgery, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Hyoun Wook Lee
- Department of Pathology, Sungkyunkwan University School of Medicine, Samsung Changwon Hospital, Changwon, Republic of Korea
| | - Young Sook Park
- Department of Physical Medicine and Rehabilitation, Sungkyunkwan University School of Medicine, Samsung Changwon Hospital, Changwon, Republic of Korea
| | - Sang Woo Park
- Department of Plastic and Reconstructive Surgery, Sungkyunkwan University School of Medicine, Samsung Changwon Hospital, Changwon, Republic of Korea
| |
Collapse
|
19
|
Low Bone Turnover Due to Hypothyroidism or Anti-Resorptive Treatment Does Not Affect Whole-Body Glucose Homeostasis in Male Mice. J Pers Med 2022; 12:jpm12091462. [PMID: 36143246 PMCID: PMC9502862 DOI: 10.3390/jpm12091462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Bone is a large and dynamic tissue and its maintenance requires high amounts of energy as old or damaged bone structures need to be replaced during the process of bone remodeling. Glucose homeostasis is an essential prerequisite for a healthy bone and vice versa, the skeleton can act as an endocrine organ on energy metabolism. We recently showed that hypothyroidism in mice leads to an almost complete arrest of bone remodeling. Here, we aimed to investigate whether the profound suppression of bone remodeling affects whole-body glucose homeostasis. To that end, male C57BL/6JRj mice were rendered hypothyroid over 4 weeks using methimazole and sodium perchlorate in the drinking water. We confirmed trabecular bone gain due to decreased bone turnover in hypothyroid mice with decreased cortical but increased vertebral bone strength. Further, we found impaired glucose handling but not insulin resistance with hypothyroidism. In hypothyroid bone, glucose uptake and expression of glucose transporter Glut4 were reduced by 44.3% and 13.9%, respectively, suggesting lower energy demands. Nevertheless, hypothyroidism led to distinct changes in glucose uptake in muscle, liver, and epididymal white adipose tissue (eWAT). Reduced glucose uptake (−30.6%) and Glut1/Glut4 transcript levels (−31.9%/−67.5%) were detected in muscle tissue. In contrast, in liver and eWAT we observed increased glucose uptake by 25.6% and 68.6%, respectively, and upregulated expression of glucose transporters with hypothyroidism. To more specifically target bone metabolism and discriminate between the skeletal and systemic effects of hypothyroidism on energy metabolism, male mice were treated with zoledronate (ZOL), a bisphosphonate, that led to decreased bone turnover, trabecular bone gain, and reduced local glucose uptake into bone (−40.4%). However, ZOL-treated mice did not display alterations of systemic glucose handling nor insulin tolerance. Despite the close mutual crosstalk of bone and glucose metabolism, in this study, we show that suppressing bone remodeling does not influence whole-body glucose homeostasis in male mice.
Collapse
|
20
|
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: 30] [Impact Index Per Article: 10.0] [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
|
21
|
Maushart CI, Senn JR, Loeliger RC, Siegenthaler J, Bur F, Fischer JGW, Betz MJ. Resting Energy Expenditure and Cold-induced Thermogenesis in Patients With Overt Hyperthyroidism. J Clin Endocrinol Metab 2022; 107:450-461. [PMID: 34570185 PMCID: PMC8764338 DOI: 10.1210/clinem/dgab706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Thyroid hormone (TH) is crucial for the adaptation to cold. OBJECTIVE To evaluate the effect of hyperthyroidism on resting energy expenditure (REE), cold-induced thermogenesis (CIT) and changes in body composition and weight. METHODS This was a prospective cohort study at the endocrine outpatient clinic of a tertiary referral center. Eighteen patients with overt hyperthyroidism were included. We measured REE during hyperthyroidism, after restoring euthyroid TH levels and after 3 months of normal thyroid function. In 14 of the 18 patients, energy expenditure (EE) was measured before and after a mild cold exposure of 2 hours and CIT was the difference between EEcold and EEwarm. Skin temperatures at 8 positions were recorded during the study visits. Body composition was assessed by dual X-ray absorption. RESULTS Free thyroxine (fT4) and free triiodothyronine (fT3) decreased significantly over time (fT4, P = .0003; fT3, P = .0001). REE corrected for lean body mass (LBM) decreased from 42 ± 6.7 kcal/24 hour/kg LBM in the hyperthyroid to 33 ± 4.4 kcal/24 hour/kg LBM (-21%, P < .0001 vs hyperthyroid) in the euthyroid state and 3 months later to 33 ± 5.2 kcal/24 hour/kg LBM (-21%, P = .0022 vs hyperthyroid, overall P < .0001). fT4 (P = .0001) and fT3 (P < 0.0001) were predictors of REE. CIT did not change from the hyperthyroid to the euthyroid state (P = .96). Hyperthyroidism led to increased skin temperature at warm ambient conditions but did not alter core body temperature, nor skin temperature after cold exposure. Weight regain and body composition were not influenced by REE and CIT during the hyperthyroid state. CONCLUSION CIT is not increased in patients with overt hyperthyroidism.
Collapse
Affiliation(s)
- Claudia I Maushart
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Jaël R Senn
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Rahel C Loeliger
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Judith Siegenthaler
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Fabienne Bur
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Jonas G W Fischer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Matthias J Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| |
Collapse
|
22
|
Senn JR, Löliger RC, Fischer JGW, Bur F, Maushart CI, Betz MJ. Acute effect of propranolol on resting energy expenditure in hyperthyroid patients. Front Endocrinol (Lausanne) 2022; 13:1026998. [PMID: 36743920 PMCID: PMC9892445 DOI: 10.3389/fendo.2022.1026998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/06/2022] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE Hyperthyroidism is a common endocrine disorder which leads to higher resting energy expenditure (REE). Increased activity of brown adipose tissue (BAT) contributes to elevated REE in hyperthyroid patients. For rapid control of hyperthyroid symptoms, the non-selective β-blocker propranolol is widely used. While, long-term treatment with propranolol reduces REE it is currently unclear whether it can also acutely diminish REE. DESIGN In the present prospective interventional trial we investigated the effect of propranolol on REE in hyperthyroid patients. METHODS Nineteen patients with overt primary hyperthyroidism were recruited from the endocrine outpatient clinic. REE was measured by indirect calorimetry before and after an acute dose of 80mg propranolol and during a control period, respectively. Additionally, skin temperature was recorded at eleven predefined locations during each study visit, vital signes and heart rate (HR) were measured before and after administration of propranolol. RESULTS Mean REE decreased slightly after acute administration of 80mg propranolol (p= 0.03) from 1639 ± 307 kcal/24h to 1594 ± 283 kcal/24h. During the control visit REE did not change significantly. HR correlated significantly with the level of free T3 (R2 = 0.38, p=0.029) free T4 (R2 = 0.39, p=0.026). HR decreased 81 ± 12 bpm to 67 ± 7.6 bpm 90 minutes after oral administration of propranolol (p<0.0001). Skin temperature did not change after propranolol intake. CONCLUSIONS In hyperthyroid patients a single dose of propranolol reduced heart rate substantially but REE diminished only marginally probably due to reduced myocardial energy consumption. Our data speak against a relevant contribution of BAT to the higher REE in hyperthyroidism. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, identifier (NCT03379181).
Collapse
Affiliation(s)
- Jaël Rut Senn
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Rahel Catherina Löliger
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Jonas Gabriel William Fischer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Fabienne Bur
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Claudia Irene Maushart
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Matthias Johannes Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- *Correspondence: Matthias Johannes Betz,
| |
Collapse
|
23
|
Cheng L, Wang J, Dai H, Duan Y, An Y, Shi L, Lv Y, Li H, Wang C, Ma Q, Li Y, Li P, Du H, Zhao B. Brown and beige adipose tissue: a novel therapeutic strategy for obesity and type 2 diabetes mellitus. Adipocyte 2021; 10:48-65. [PMID: 33403891 PMCID: PMC7801117 DOI: 10.1080/21623945.2020.1870060] [Citation(s) in RCA: 214] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mammalian adipose tissue can be divided into two major types, namely, white adipose tissue (WAT) and brown adipose tissue (BAT). According to classical view, the main function of WAT is to store excess energy in the form of triglycerides, while BAT is a thermogenic tissue that acts a pivotal part in maintaining the core body temperature. White adipocytes display high plasticity and can transdifferentiate into beige adipocytes which have many similar morphological and functional properties with brown adipocytes under the stimulations of exercise, cold exposure and other factors. This phenomenon is also known as 'browning of WAT'. In addition to transdifferentiation, beige adipocytes can also come from de novo differentiation from tissue-resident progenitors. Activating BAT and inducing browning of WAT can accelerate the intake of glycolipids and reduce the insulin secretion requirement, which may be a new strategy to improve glycolipids metabolism and insulin resistance of obese and type 2 diabetes mellitus (T2DM) patients. This review mainly discusses the significance of brown and beige adipose tissues in the treatment of obesity and T2DM, and focuses on the effect of the browning agent on obesity and T2DM, which provides a brand-new theoretical reference for the prevention and treatment of obesity and T2DM.
Collapse
Affiliation(s)
- Long Cheng
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing China
| | - Jingkang Wang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing China
| | - Hongyu Dai
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing China
| | - Yuhui Duan
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing China
| | - Yongcheng An
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Lu Shi
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing China
| | - Yinglan Lv
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing China
| | - Huimin Li
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing China
| | - Chen Wang
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Quantao Ma
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing China
| | - Yaqi Li
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing China
| | - Pengfei Li
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing China
| | - Haifeng Du
- The Third Municipal Hospital of Chengde, Chengde, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing China
| |
Collapse
|
24
|
Capelli V, Grijota-Martínez C, Dragano NRV, Rial-Pensado E, Fernø J, Nogueiras R, Mittag J, Diéguez C, López M. Orally Induced Hyperthyroidism Regulates Hypothalamic AMP-Activated Protein Kinase. Nutrients 2021; 13:nu13124204. [PMID: 34959756 PMCID: PMC8708331 DOI: 10.3390/nu13124204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022] Open
Abstract
Besides their direct effects on peripheral metabolic tissues, thyroid hormones (TH) act on the hypothalamus to modulate energy homeostasis. However, since most of the hypothalamic actions of TH have been addressed in studies with direct central administration, the estimation of the relative contribution of the central vs. peripheral effects in physiologic conditions of peripheral release (or administration) of TH remains unclear. In this study we used two different models of peripherally induced hyperthyroidism (i.e., T4 and T3 oral administration) to assess and compare the serum and hypothalamic TH status and relate them to the metabolic effects of the treatment. Peripheral TH treatment affected feeding behavior, overall growth, core body temperature, body composition, brown adipose tissue (BAT) morphology and uncoupling protein 1 (UCP1) levels and metabolic activity, white adipose tissue (WAT) browning and liver metabolism. This resulted in an increased overall uncoupling capacity and a shift of the lipid metabolism from WAT accumulation to BAT fueling. Both peripheral treatment protocols induced significant changes in TH concentrations within the hypothalamus, with T3 eliciting a downregulation of hypothalamic AMP-activated protein kinase (AMPK), supporting the existence of a central action of peripheral TH. Altogether, these data suggest that peripherally administered TH modulate energy balance by various mechanisms; they also provide a unifying vision of the centrally mediated and the direct local metabolic effect of TH in the context of hyperthyroidism.
Collapse
Affiliation(s)
- Valentina Capelli
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (V.C.); (N.R.V.D.); (E.R.-P.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Madrid, Spain
- Unit of Internal Medicine and Endocrinology, Istituti Clinici Scientifici Maugeri, Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Carmen Grijota-Martínez
- Department of Cell Biology, Faculty of Biology, Complutense University, 28040 Madrid, Spain;
| | - Nathalia R. V. Dragano
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (V.C.); (N.R.V.D.); (E.R.-P.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Madrid, Spain
| | - Eval Rial-Pensado
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (V.C.); (N.R.V.D.); (E.R.-P.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Madrid, Spain
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway;
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (V.C.); (N.R.V.D.); (E.R.-P.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Madrid, Spain
| | - Jens Mittag
- Institute for Endocrinology and Diabetes—Molecular Endocrinology, Center of Brain Behavior and Metabolism CBBM, University of Lübeck, 23562 Lübeck, Germany;
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (V.C.); (N.R.V.D.); (E.R.-P.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Madrid, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; (V.C.); (N.R.V.D.); (E.R.-P.); (R.N.); (C.D.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Madrid, Spain
- Correspondence: ; Tel.: +34-881815420
| |
Collapse
|
25
|
Weiner J, Roth L, Kranz M, Brust P, Boelen A, Klöting N, Heiker JT, Blüher M, Tönjes A, Pfluger PT, Stumvoll M, Mittag J, Krause K. Leptin counteracts hypothermia in hypothyroidism through its pyrexic effects and by stabilizing serum thyroid hormone levels. Mol Metab 2021; 54:101348. [PMID: 34610354 PMCID: PMC8556519 DOI: 10.1016/j.molmet.2021.101348] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 11/30/2022] Open
Abstract
Objective Thyroid hormones (TH) are essential for the homeostatic control of energy metabolism and the regulation of body temperature. The hypothalamic–pituitary–thyroid (HPT) axis is regulated by negative feedback mechanisms, ensuring that TH levels are maintained at a constant level. However, the feedback mechanisms underlying the resetting of the HPT axis regulation in the control of body temperature are still not fully understood. Here, we aimed to determine the thermoregulatory response in hypothyroid mice to different environmental temperatures and the underlying mechanisms. Methods Distinct thermogenic challenges were induced in hypothyroid female C57BL/6N and leptin-deficient ob/ob mice through housing at either room temperature or thermoneutrality. The thermogenic and metabolic effects were analyzed through metabolic chambers, 18F-FDG-PET/MRI, infrared thermography, metabolic profiling, histology, gene expression and Western blot analysis. Results In hypothyroid mice maintained at room temperature, high leptin serum levels induce a pyrexic effect leading to the stabilization of body temperature through brown adipose tissue thermogenesis and white adipose tissue browning. Housing at thermoneutrality leads to the normalization of leptin levels and a reduction of the central temperature set point, resulting in decreased thermogenesis in brown and white adipose tissue and skeletal muscle and a significant decline in body temperature. Furthermore, anapyrexia in hypothyroid leptin-deficient ob/ob mice indicates that besides its pyrexic actions, leptin exerts a stimulatory effect on the HPT axis to stabilize the remaining TH serum levels in hypothyroid mice. Conclusion This study led to the identification of a previously unknown endocrine loop in which leptin acts in concert with the HPT axis to stabilize body temperature in hypothyroid mice. Thyroid hormones are essential for the regulation of body temperature. Thyroid hormone-deficient (hypothyroid) mice show distinct leptin serum concentrations in response to changes in ambient housing temperature. High leptin serum levels confer a stimulatory effect on the hypothalamic-pituitary-thyroid axis. High leptin serum level prevents fall in body temperature in hypothyroid mice at room temperature through its pyrexic effects.
Collapse
Affiliation(s)
- Juliane Weiner
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Lisa Roth
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Mathias Kranz
- University Hospital of North Norway, Tromsø, Norway; Helmholtz-Zentrum Dresden-Rossendorf, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Anita Boelen
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Nora Klöting
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - John T Heiker
- Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - Anke Tönjes
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Paul T Pfluger
- Helmholtz Zentrum München, Research Unit NeuroBiology of Diabetes, Neuherberg, Germany; Technical University of Munich (TUM), TUM School of Medicine, NeuroBiology of Diabetes, Munich, Germany
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - Jens Mittag
- Institute for Endocrinology & Diabetes/CBBM, University of Lübeck, Lübeck, Germany
| | - Kerstin Krause
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
26
|
Walczak K, Sieminska L. Obesity and Thyroid Axis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18189434. [PMID: 34574358 PMCID: PMC8467528 DOI: 10.3390/ijerph18189434] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/28/2021] [Accepted: 09/03/2021] [Indexed: 12/19/2022]
Abstract
Development of obesity is primarily the result of imbalance between energy intake and energy expenditure. Thyroid hormones influence energy expenditure by regulating cellular respiration and thermogenesis and by determining resting metabolic rate. Triiodothyronine influences lipid turnover in adipocytes and impacts appetite regulation through the central nervous system, mainly the hypothalamus. Thyroid-stimulating hormone may also influence thermogenesis, suppress appetite and regulate lipid storage through lipolysis and lipogenesis control. Subclinical hypothyroidism may induce changes in basal metabolic rate with subsequent increase in BMI, but obesity can also affect thyroid function via several mechanisms such as lipotoxicity and changes in adipokines and inflammatory cytokine secretion. The present study investigated the complex and mutual relationships between the thyroid axis and adiposity.
Collapse
Affiliation(s)
- Krzysztof Walczak
- Department of Thoracic Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland;
| | - Lucyna Sieminska
- Department of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland
- Correspondence:
| |
Collapse
|
27
|
Capelli V, Diéguez C, Mittag J, López M. Thyroid wars: the rise of central actions. Trends Endocrinol Metab 2021; 32:659-671. [PMID: 34294513 DOI: 10.1016/j.tem.2021.05.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/30/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022]
Abstract
In the field of thyroid hormone (TH) action on energy balance, huge advances have been achieved in the past decade, from human, animal, and in vitro studies. A key achievement was the demonstration of the TH 'central' metabolic action, which was recently discovered in rodent models and challenged the previous 'peripheral' paradigm. In this opinion, we dissect and try to unify the two paradigms, from analyzing the respective bench models to extrapolating the possible translational bedside implications.
Collapse
Affiliation(s)
- Valentina Capelli
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Jens Mittag
- University of Lübeck, Institute for Endocrinology and Diabetes, Center of Brain Behavior and Metabolism (CBBM), Lübeck, Germany.
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain.
| |
Collapse
|
28
|
Sun L, Goh HJ, Verma S, Govindharajulu P, Sadananthan SA, Michael N, Jadegoud Y, Henry CJ, Velan SS, Yeo PS, Lee Y, Lim BSP, Liew H, Chew CK, Quek TPL, Abdul Shakoor SAKK, Hoi WH, Chan SP, Chew DE, Dalan R, Leow MKS. Metabolic effects of brown fat in transitioning from hyperthyroidism to euthyroidism. Eur J Endocrinol 2021; 185:553-563. [PMID: 34342595 PMCID: PMC8428075 DOI: 10.1530/eje-21-0366] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/03/2021] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Brown adipose tissue (BAT) controls metabolic rate through thermogenesis. As its regulatory factors during the transition from hyperthyroidism to euthyroidism are not well established, our study investigated the relationships between supraclavicular brown adipose tissue (sBAT) activity and physiological/metabolic changes with changes in thyroid status. DESIGN Participants with newly diagnosed Graves' disease were recruited. A thionamide antithyroid drug (ATD) such as carbimazole (CMZ) or thiamazole (TMZ) was prescribed in every case. All underwent energy expenditure (EE) measurement and supraclavicular infrared thermography (IRT) within a chamber calorimeter, as well as 18F-fluorodeoxyglucose (18F-FDG) positron-emission tomography/magnetic resonance (PET/MR) imaging scanning, with clinical and biochemical parameters measured during hyperthyroidism and repeated in early euthyroidism. PET sBAT mean/maximum standardized uptake value (SUV mean/max), MR supraclavicular fat fraction (sFF) and mean temperature (Tscv) quantified sBAT activity. RESULTS Twenty-one (16 female/5 male) participants aged 39.5 ± 2.5 years completed the study. The average duration to attain euthyroidism was 28.6 ± 2.3 weeks. Eight participants were BAT-positive while 13 were BAT-negative. sFF increased with euthyroidism (72.3 ± 1.4% to 76.8 ± 1.4%; P < 0.01), but no changes were observed in PET SUV mean and Tscv. Significant changes in serum-free triiodothyronine (FT3) levels were related to BAT status (interaction P value = 0.04). FT3 concentration at hyperthyroid state was positively associated with sBAT PET SUV mean (r = 0.58, P = 0.01) and resting metabolic rate (RMR) (P < 0.01). CONCLUSION Hyperthyroidism does not consistently lead to a detectable increase in BAT activity. FT3 reduction during the transition to euthyroidism correlated with BAT activity.
Collapse
Affiliation(s)
- Lijuan Sun
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Hui Jen Goh
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sanjay Verma
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Priya Govindharajulu
- Singapore Institute of Food and Biotechnology Innovation, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Suresh Anand Sadananthan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Navin Michael
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Yaligar Jadegoud
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Christiani Jeyakumar Henry
- Singapore Institute of Food and Biotechnology Innovation, Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine
| | - S Sendhil Velan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research (A*STAR), Singapore
- Departments of Physiology & Medicine, National University of Singapore (NUS), Singapore
| | - Pei Shan Yeo
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital (TTSH), Singapore
| | - Yingshan Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital (TTSH), Singapore
| | - Brenda Su Ping Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital (TTSH), Singapore
| | - Huiling Liew
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital (TTSH), Singapore
| | - Chee Kian Chew
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital (TTSH), Singapore
| | - Timothy Peng Lim Quek
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital (TTSH), Singapore
| | - Shaikh A K K Abdul Shakoor
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital (TTSH), Singapore
| | - Wai Han Hoi
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital (TTSH), Singapore
| | - Siew Pang Chan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Daniel Ek Chew
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital (TTSH), Singapore
| | - Rinkoo Dalan
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital (TTSH), Singapore
| | - Melvin Khee Shing Leow
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital (TTSH), Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
- Correspondence should be addressed to M K Leow Email
| |
Collapse
|
29
|
Hoffmann A, Ebert T, Hankir MK, Flehmig G, Klöting N, Jessnitzer B, Lössner U, Stumvoll M, Blüher M, Fasshauer M, Tönjes A, Miehle K, Kralisch S. Leptin Improves Parameters of Brown Adipose Tissue Thermogenesis in Lipodystrophic Mice. Nutrients 2021; 13:2499. [PMID: 34444659 PMCID: PMC8399124 DOI: 10.3390/nu13082499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 12/18/2022] Open
Abstract
Lipodystrophy syndromes (LD) are a heterogeneous group of very rare congenital or acquired disorders characterized by a generalized or partial lack of adipose tissue. They are strongly associated with severe metabolic dysfunction due to ectopic fat accumulation in the liver and other organs and the dysregulation of several key adipokines, including leptin. Treatment with leptin or its analogues is therefore sufficient to reverse some of the metabolic symptoms of LD in patients and in mouse models through distinct mechanisms. Brown adipose tissue (BAT) thermogenesis has emerged as an important regulator of systemic metabolism in rodents and in humans, but it is poorly understood how leptin impacts BAT in LD. Here, we show in transgenic C57Bl/6 mice overexpressing sterol regulatory element-binding protein 1c in adipose tissue (Tg (aP2-nSREBP1c)), an established model of congenital LD, that daily subcutaneous administration of 3 mg/kg leptin for 6 to 8 weeks increases body temperature without affecting food intake or body weight. This is associated with increased protein expression of the thermogenic molecule uncoupling protein 1 (UCP1) and the sympathetic nerve marker tyrosine hydroxylase (TH) in BAT. These findings suggest that leptin treatment in LD stimulates BAT thermogenesis through sympathetic nerves, which might contribute to some of its metabolic benefits by providing a healthy reservoir for excess circulating nutrients.
Collapse
Affiliation(s)
- Annett Hoffmann
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04109 Leipzig, Germany; (T.E.); (G.F.); (B.J.); (U.L.); (M.S.); (M.B.); (A.T.); (K.M.); (S.K.)
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080 Würzburg, Germany;
| | - Thomas Ebert
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04109 Leipzig, Germany; (T.E.); (G.F.); (B.J.); (U.L.); (M.S.); (M.B.); (A.T.); (K.M.); (S.K.)
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17177 Solna, Sweden
| | - Mohammed K. Hankir
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080 Würzburg, Germany;
| | - Gesine Flehmig
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04109 Leipzig, Germany; (T.E.); (G.F.); (B.J.); (U.L.); (M.S.); (M.B.); (A.T.); (K.M.); (S.K.)
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig, 04109 Leipzig, Germany;
| | - Beate Jessnitzer
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04109 Leipzig, Germany; (T.E.); (G.F.); (B.J.); (U.L.); (M.S.); (M.B.); (A.T.); (K.M.); (S.K.)
| | - Ulrike Lössner
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04109 Leipzig, Germany; (T.E.); (G.F.); (B.J.); (U.L.); (M.S.); (M.B.); (A.T.); (K.M.); (S.K.)
| | - Michael Stumvoll
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04109 Leipzig, Germany; (T.E.); (G.F.); (B.J.); (U.L.); (M.S.); (M.B.); (A.T.); (K.M.); (S.K.)
| | - Matthias Blüher
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04109 Leipzig, Germany; (T.E.); (G.F.); (B.J.); (U.L.); (M.S.); (M.B.); (A.T.); (K.M.); (S.K.)
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig, 04109 Leipzig, Germany;
| | - Mathias Fasshauer
- Institute of Nutritional Science, Justus-Liebig-University, 35390 Giessen, Germany;
| | - Anke Tönjes
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04109 Leipzig, Germany; (T.E.); (G.F.); (B.J.); (U.L.); (M.S.); (M.B.); (A.T.); (K.M.); (S.K.)
| | - Konstanze Miehle
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04109 Leipzig, Germany; (T.E.); (G.F.); (B.J.); (U.L.); (M.S.); (M.B.); (A.T.); (K.M.); (S.K.)
| | - Susan Kralisch
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04109 Leipzig, Germany; (T.E.); (G.F.); (B.J.); (U.L.); (M.S.); (M.B.); (A.T.); (K.M.); (S.K.)
| |
Collapse
|
30
|
Bao MH, Xu XM, Huo DL, Cao J, Zhao ZJ. The effect of aggression II: Acclimation to a high ambient temperature reduces territorial aggression in male striped hamsters (Cricetulus barabensis). Horm Behav 2021; 132:104993. [PMID: 33991799 DOI: 10.1016/j.yhbeh.2021.104993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/01/2022]
Abstract
Thyroid hormones have a profound influence on development, cellular differentiation and metabolism, and are also suspected of playing a role in aggression. We measured territorial aggression, body temperature (Tb) and serum thyroid hormones levels of male striped hamsters (Cricetulus barabensis) acclimated to either cold (5 °C), cool (21 °C) or hot (34 °C) ambient temperatures. The effects of methimazole on territorial aggression, food intake, metabolic rate and serum thyroid hormone levels, were also examined. Territorial aggression was significantly lower in male hamsters acclimated to the hot temperature compared to those acclimated to the cool or cold temperatures. Tb significantly increased during aggressive territorial interactions with intruders but did not significantly differ among the three temperature treatments. Serum T3, T4 and cortisol levels of hamsters acclimated to 34 °C were significantly lower than those acclimated to 21 °C. In addition to significantly reducing territorial aggression, treatment with methimazole also significantly reduced serum T3 and T4 levels, Tb and metabolic rate. These results suggest that exposure to high temperatures reduces the capacity of hamsters to dissipate heat causing them to lower their metabolic rate, which, in turn, causes them to reduce territorial aggression to prevent hyperthermia. The lower metabolic rate mediated by down-regulated thyroid hormones inhibits territorial aggression and could thereby determine the outcome of territorial conflicts.
Collapse
Affiliation(s)
- Meng-Huan Bao
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Xiao-Ming Xu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Da-Liang Huo
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Jing Cao
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Zhi-Jun Zhao
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
31
|
Zekri Y, Flamant F, Gauthier K. Central vs. Peripheral Action of Thyroid Hormone in Adaptive Thermogenesis: A Burning Topic. Cells 2021; 10:1327. [PMID: 34071979 PMCID: PMC8229489 DOI: 10.3390/cells10061327] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/18/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Thyroid hormones (TH) contribute to the control of adaptive thermogenesis, which is associated with both higher energy expenditure and lower body mass index. While it was clearly established that TH act directly in the target tissues to fulfill its metabolic activities, some studies have rather suggested that TH act in the hypothalamus to control these processes. This paradigm shift has subjected the topic to intense debates. This review aims to recapitulate how TH control adaptive thermogenesis and to what extent the brain is involved in this process. This is of crucial importance for the design of new pharmacological agents that would take advantage of the TH metabolic properties.
Collapse
Affiliation(s)
- Yanis Zekri
- Institut de Génomique Fonctionnelle de Lyon, Univ Lyon, CNRS UMR 5242, INRAE USC 1370 École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, 46 allée d’Italie, 69007 Lyon, France; (F.F.); (K.G.)
| | | | | |
Collapse
|
32
|
Roth CL, Molica F, Kwak BR. Browning of White Adipose Tissue as a Therapeutic Tool in the Fight against Atherosclerosis. Metabolites 2021; 11:319. [PMID: 34069148 PMCID: PMC8156962 DOI: 10.3390/metabo11050319] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/05/2021] [Accepted: 05/13/2021] [Indexed: 02/07/2023] Open
Abstract
Despite continuous medical advances, atherosclerosis remains the prime cause of mortality worldwide. Emerging findings on brown and beige adipocytes highlighted that these fat cells share the specific ability of non-shivering thermogenesis due to the expression of uncoupling protein 1. Brown fat is established during embryogenesis, and beige cells emerge from white adipose tissue exposed to specific stimuli like cold exposure into a process called browning. The consecutive energy expenditure of both thermogenic adipose tissues has shown therapeutic potential in metabolic disorders like obesity and diabetes. The latest data suggest promising effects on atherosclerosis development as well. Upon cold exposure, mice and humans have a physiological increase in brown adipose tissue activation and browning of white adipocytes is promoted. The use of drugs like β3-adrenergic agonists in murine models induces similar effects. With respect to atheroprotection, thermogenic adipose tissue activation has beneficial outcomes in mice by decreasing plasma triglycerides, total cholesterol and low-density lipoproteins, by increasing high-density lipoproteins, and by inducing secretion of atheroprotective adipokines. Atheroprotective effects involve an unaffected hepatic clearance. Latest clinical data tend to find thinner atherosclerotic lesions in patients with higher brown adipose tissue activity. Strategies for preserving healthy arteries are a major concern for public health.
Collapse
Affiliation(s)
| | - Filippo Molica
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland; (C.L.R.); (B.R.K.)
| | | |
Collapse
|
33
|
Volke L, Krause K. Effect of Thyroid Hormones on Adipose Tissue Flexibility. Eur Thyroid J 2021; 10:1-9. [PMID: 33777816 PMCID: PMC7983599 DOI: 10.1159/000508483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/05/2020] [Indexed: 12/18/2022] Open
Abstract
The recruitment and activation of energy-consuming brown adipocytes is currently considered as potential therapeutic approach to combat obesity. Thyroid hormones (TH) significantly contribute to full thermogenic capacity of brown adipocytes. A number of recent studies suggest that TH also induce the recruitment of brown adipocytes in white adipose depots, a process known as browning. In this review, we will summarize underlying mechanisms by which TH mediate brown adipose tissue activity and white adipose tissue browning. Furthermore, we will discuss the relevance of TH-induced white adipose tissue browning for thermoregulation.
Collapse
Affiliation(s)
- Lisa Volke
- Medical Department III-Endocrinology, Nephrology, and Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Kerstin Krause
- Medical Department III-Endocrinology, Nephrology, and Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Medicine, University of Leipzig, Leipzig, Germany
- *Kerstin Krause, Department of Medicine, University of Leipzig, Liebigstrasse 21, DE–04103 Leipzig (Germany),
| |
Collapse
|
34
|
Sentis SC, Oelkrug R, Mittag J. Thyroid hormones in the regulation of brown adipose tissue thermogenesis. Endocr Connect 2021; 10:R106-R115. [PMID: 33491659 PMCID: PMC7983518 DOI: 10.1530/ec-20-0562] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/20/2021] [Indexed: 12/15/2022]
Abstract
A normal thyroid status is crucial for body temperature homeostasis, as thyroid hormone regulates both heat loss and conservation as well as heat production in the thermogenic tissues. Brown adipose tissue (BAT) is the major site of non-shivering thermogenesis and an important target of thyroid hormone action. Thyroid hormone not only regulates the tissue's sensitivity to sympathetic stimulation by norepinephrine but also the expression of uncoupling protein 1, the key driver of BAT thermogenesis. Vice versa, sympathetic stimulation of BAT triggers the expression of deiodinase type II, an enzyme that enhances local thyroid hormone availability and signaling. This review summarizes the current knowledge on how thyroid hormone controls BAT thermogenesis, aiming to dissect the direct actions of the hormone in BAT and its indirect actions via the CNS, browning of white adipose tissue or heat loss over body surfaces. Of particular relevance is the apparent dose dependency of the observed effects, as we find that minor or moderate changes in thyroid hormone levels often have different effects as compared to high pharmacological doses. Moreover, we conclude that the more recent findings require a reevaluation of older studies, as key aspects such as heat loss or central BAT activation may not have received the necessary attention during the interpretation of these early findings. Finally, we provide a list of what we believe are the most relevant questions in the field that to date are still enigmatic and require further studies.
Collapse
Affiliation(s)
- Sarah Christine Sentis
- Institute for Endocrinology and Diabetes, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Rebecca Oelkrug
- Institute for Endocrinology and Diabetes, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Jens Mittag
- Institute for Endocrinology and Diabetes, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- Correspondence should be addressed to J Mittag:
| |
Collapse
|
35
|
Li GL, Ping J, Chen HJ, Zhang WX, Fan J, Peng DS, Zhang L, Yan YE. Maternal nicotine exposure impairs brown adipose tissue via AMPK-SIRT1-PGC-1α signals in male offspring. Life Sci 2021; 264:118695. [PMID: 33130079 DOI: 10.1016/j.lfs.2020.118695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
AIMS Maternal nicotine exposure during pregnancy and lactation is associated with obesity in offspring. Brown adipose tissue (BAT) is correlated with energy metabolism and obesity. In this study, we explored the mechanism of maternal nicotine exposure on BAT changes in male offspring. MAIN METHODS Pregnant rats were randomly assigned to nicotine (1.0 mg/kg twice per day, subcutaneous administration) or control groups. In vitro, C3H10T1/2 cells were induced to differentiate into mature brown adipocytes, and 0-50 μM nicotine was given to C3H10T1/2 cells during the differentiation process. KEY FINDINGS Nicotine-exposed males had white-like adipocytes and abnormal mitochondria structure in iBAT at 26 weeks. The expression of mitochondrial genes, UCP1 and AMPK-SIRT1-PGC-1α pathway were downregulated in the nicotine group at 26 weeks rather than 4 weeks. In vitro, 50 μM nicotine decreased the expression of mitochondrial genes, UCP1 and AMPK-SIRT1-PGC-1α pathway in brown adipocytes. SIGNIFICANCE Maternal nicotine exposure showed the "programming" effect on the decreased brown-like phenotype in BAT of adult male offspring via downregulating AMPK-SIRT1-PGC-1α pathway. This impairment of BAT may be a potential mechanism of nicotine-induced obesity in male offspring.
Collapse
Affiliation(s)
- Gai-Ling Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Jie Ping
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hui-Jian Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Wan-Xia Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Jie Fan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Dang-Sheng Peng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Li Zhang
- Demonstration Center for Experimental Basic Medicine Education of Wuhan University, China
| | - You-E Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
| |
Collapse
|
36
|
Frare C, Williams CT, Drew KL. Thermoregulation in hibernating mammals: The role of the "thyroid hormones system". Mol Cell Endocrinol 2021; 519:111054. [PMID: 33035626 PMCID: PMC8091518 DOI: 10.1016/j.mce.2020.111054] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 07/15/2020] [Accepted: 10/04/2020] [Indexed: 12/19/2022]
Abstract
Hibernation is a unique evolutionary adaptation to conserve energy. During the pre-hibernation (i.e. fall) season, a progressive decline in core body temperature and further decrease in metabolism underlie a seasonal modulation in thermoregulation. The onset of hibernation requires marked changes in thermoregulatory attributes including adjustment in body temperature and tissue specific increases in thermogenic capacity. The hibernation season is characterized by a regulated suppression in thermogenesis allowing the onset of torpor interrupted by periodic activation of thermogenesis to sustain interbout arousals. Thyroid hormones are known to regulate both body temperature and metabolism, and for this reason, the hypothalamic-pituitary-thyroid axis and thyroid hormones have been investigated as modulators of thermogenesis in the phenomenon of hibernation, but the mechanisms remain poorly understood. In this review, we present an overview of what is known about the thermogenic roles of thyroid hormones in hibernating species across seasons and within the hibernating season (torpor-interbout arousal cycle). Overall, the hypothalamic-pituitary-thyroid axis and thyroid hormones play a role in the pre-hibernation season to enhance thermogenic capacity. During hibernation, thermogenesis is attenuated at the level of sympathetic premotor neurons within the raphe pallidus and by deiodinase expression in the hypothalamus. Further, as recent work highlights the direct effect of thyroid hormones within the central nervous system in activating thermogenesis, we speculate how similar mechanisms may occur in hibernating species to modulate thermogenesis across seasons and to sustain interbout arousals. However, further experiments are needed to elucidate the role of thyroid hormones in hibernation, moving towards the understanding that thyroid hormones metabolism, transport and availability within tissues may be the most telling indicator of thyroid status.
Collapse
Affiliation(s)
- C Frare
- Department of Chemistry and Biochemistry University of Alaska Fairbanks, Fairbanks, AK, 99775, USA; Institute of Arctic Biology, Center for Transformative Research in Metabolism, University of Alaska Fairbanks 2140 Koyukuk Drive, Fairbanks, AK, 99775, USA
| | - Cory T Williams
- Institute of Arctic Biology, Center for Transformative Research in Metabolism, University of Alaska Fairbanks 2140 Koyukuk Drive, Fairbanks, AK, 99775, USA; Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK, 99775, USA
| | - Kelly L Drew
- Department of Chemistry and Biochemistry University of Alaska Fairbanks, Fairbanks, AK, 99775, USA; Institute of Arctic Biology, Center for Transformative Research in Metabolism, University of Alaska Fairbanks 2140 Koyukuk Drive, Fairbanks, AK, 99775, USA.
| |
Collapse
|
37
|
Gauthier BR, Sola‐García A, Cáliz‐Molina MÁ, Lorenzo PI, Cobo‐Vuilleumier N, Capilla‐González V, Martin‐Montalvo A. Thyroid hormones in diabetes, cancer, and aging. Aging Cell 2020; 19:e13260. [PMID: 33048427 PMCID: PMC7681062 DOI: 10.1111/acel.13260] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/27/2020] [Accepted: 09/13/2020] [Indexed: 12/18/2022] Open
Abstract
Thyroid function is central in the control of physiological and pathophysiological processes. Studies in animal models and human research have determined that thyroid hormones modulate cellular processes relevant for aging and for the majority of age‐related diseases. While several studies have associated mild reductions on thyroid hormone function with exceptional longevity in animals and humans, alterations in thyroid hormones are serious medical conditions associated with unhealthy aging and premature death. Moreover, both hyperthyroidism and hypothyroidism have been associated with the development of certain types of diabetes and cancers, indicating a great complexity of the molecular mechanisms controlled by thyroid hormones. In this review, we describe the latest findings in thyroid hormone research in the field of aging, diabetes, and cancer, with a special focus on hepatocellular carcinomas. While aging studies indicate that the direct modulation of thyroid hormones is not a viable strategy to promote healthy aging or longevity and the development of thyromimetics is challenging due to inefficacy and potential toxicity, we argue that interventions based on the use of modulators of thyroid hormone function might provide therapeutic benefit in certain types of diabetes and cancers.
Collapse
Affiliation(s)
- Benoit R. Gauthier
- Department of Cell Therapy and Regeneration Andalusian Center for Molecular Biology and Regenerative Medicine‐CABIMER Junta de Andalucía‐University of Pablo de Olavide‐University of Seville‐CSIC Seville Spain
- Biomedical Research Network on Diabetes and Related Metabolic Diseases‐CIBERDEM Instituto de Salud Carlos III Madrid Spain
| | - Alejandro Sola‐García
- Department of Cell Therapy and Regeneration Andalusian Center for Molecular Biology and Regenerative Medicine‐CABIMER Junta de Andalucía‐University of Pablo de Olavide‐University of Seville‐CSIC Seville Spain
| | - María Ángeles Cáliz‐Molina
- Department of Cell Therapy and Regeneration Andalusian Center for Molecular Biology and Regenerative Medicine‐CABIMER Junta de Andalucía‐University of Pablo de Olavide‐University of Seville‐CSIC Seville Spain
| | - Petra Isabel Lorenzo
- Department of Cell Therapy and Regeneration Andalusian Center for Molecular Biology and Regenerative Medicine‐CABIMER Junta de Andalucía‐University of Pablo de Olavide‐University of Seville‐CSIC Seville Spain
| | - Nadia Cobo‐Vuilleumier
- Department of Cell Therapy and Regeneration Andalusian Center for Molecular Biology and Regenerative Medicine‐CABIMER Junta de Andalucía‐University of Pablo de Olavide‐University of Seville‐CSIC Seville Spain
| | - Vivian Capilla‐González
- Department of Cell Therapy and Regeneration Andalusian Center for Molecular Biology and Regenerative Medicine‐CABIMER Junta de Andalucía‐University of Pablo de Olavide‐University of Seville‐CSIC Seville Spain
| | - Alejandro Martin‐Montalvo
- Department of Cell Therapy and Regeneration Andalusian Center for Molecular Biology and Regenerative Medicine‐CABIMER Junta de Andalucía‐University of Pablo de Olavide‐University of Seville‐CSIC Seville Spain
| |
Collapse
|
38
|
The paradoxical lean phenotype of hypothyroid mice is marked by increased adaptive thermogenesis in the skeletal muscle. Proc Natl Acad Sci U S A 2020; 117:22544-22551. [PMID: 32826330 DOI: 10.1073/pnas.2008919117] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Obesity is a major health problem worldwide, given its growing incidence and its association with a variety of comorbidities. Weight gain results from an increase in energy intake without a concomitant increase in energy expenditure. To combat the obesity epidemic, many studies have focused on the pathways underlying satiety and hunger signaling, while other studies have concentrated on the mechanisms involved in energy expenditure, most notably adaptive thermogenesis. Hypothyroidism in humans is typically associated with a decreased basal metabolic rate, lower energy expenditure, and weight gain. However, hypothyroid mouse models have been reported to have a leaner phenotype than euthyroid controls. To elucidate the mechanism underlying this phenomenon, we used a drug-free mouse model of hypothyroidism: mice lacking the sodium/iodide symporter (NIS), the plasma membrane protein that mediates active iodide uptake in the thyroid. In addition to being leaner than euthyroid mice, owing in part to reduced food intake, these hypothyroid mice show signs of compensatory up-regulation of the skeletal-muscle adaptive thermogenic marker sarcolipin, with an associated increase in fatty acid oxidation (FAO). Neither catecholamines nor thyroid-stimulating hormone (TSH) are responsible for sarcolipin expression or FAO stimulation; rather, thyroid hormones are likely to negatively regulate both processes in skeletal muscle. Our findings indicate that hypothyroidism in mice results in a variety of metabolic changes, which collectively lead to a leaner phenotype. A deeper understanding of these changes may make it possible to develop new strategies against obesity.
Collapse
|
39
|
Yau WW, Yen PM. Thermogenesis in Adipose Tissue Activated by Thyroid Hormone. Int J Mol Sci 2020; 21:ijms21083020. [PMID: 32344721 PMCID: PMC7215895 DOI: 10.3390/ijms21083020] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023] Open
Abstract
Thermogenesis is the production of heat that occurs in all warm-blooded animals. During cold exposure, there is obligatory thermogenesis derived from body metabolism as well as adaptive thermogenesis through shivering and non-shivering mechanisms. The latter mainly occurs in brown adipose tissue (BAT) and muscle; however, white adipose tissue (WAT) also can undergo browning via adrenergic stimulation to acquire thermogenic potential. Thyroid hormone (TH) also exerts profound effects on thermoregulation, as decreased body temperature and increased body temperature occur during hypothyroidism and hyperthyroidism, respectively. We have termed the TH-mediated thermogenesis under thermoneutral conditions “activated” thermogenesis. TH acts on the brown and/or white adipose tissues to induce uncoupled respiration through the induction of the uncoupling protein (Ucp1) to generate heat. TH acts centrally to activate the BAT and browning through the sympathetic nervous system. However, recent studies also show that TH acts peripherally on the BAT to directly stimulate Ucp1 expression and thermogenesis through an autophagy-dependent mechanism. Additionally, THs can exert Ucp1-independent effects on thermogenesis, most likely through activation of exothermic metabolic pathways. This review summarizes thermogenic effects of THs on adipose tissues.
Collapse
Affiliation(s)
- Winifred W Yau
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke NUS Medical School, Singapore 169857, Singapore
| | - Paul M Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke NUS Medical School, Singapore 169857, Singapore
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27708, USA
| |
Collapse
|
40
|
Ochsner SA, McKenna NJ. No Dataset Left Behind: Mechanistic Insights into Thyroid Receptor Signaling Through Transcriptomic Consensome Meta-Analysis. Thyroid 2020; 30:621-639. [PMID: 31910096 PMCID: PMC7187985 DOI: 10.1089/thy.2019.0307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background: Discovery-scale omics datasets relevant to thyroid receptors (TRs) and their physiological and synthetic bioactive small-molecule ligands allow for genome-wide interrogation of TR-regulated genes. These datasets have considerable collective value as a reference resource to allow researchers to routinely generate hypotheses addressing the mechanisms underlying the cell biology and physiology of TR signaling in normal and disease states. Methods: Here, we searched the Gene Expression Omnibus database to identify a population of publicly archived transcriptomic datasets involving genetic or pharmacological manipulation of either TR isoform in a mouse tissue or cell line. After initial quality control, samples were organized into contrasts (experiments), and transcript differential expression values and associated measures of significance were generated and committed to a consensome (for consensus omics) meta-analysis pipeline. To gain insight into tissue-selective functions of TRs, we generated liver- and central nervous system (CNS)-specific consensomes and identified evidence for genes that were selectively responsive to TR signaling in each organ. Results: The TR transcriptomic consensome ranks genes based on the frequency of their significant differential expression over the entire group of experiments. The TR consensome assigns elevated rankings both to known TR-regulated genes and to genes previously uncharacterized as TR-regulated, which shed mechanistic light on known cellular and physiological roles of TR signaling in different organs. We identify evidence for unreported genomic targets of TR signaling for which it exhibits strikingly distinct regulatory preferences in the liver and CNS. Moreover, the intersection of the TR consensome with consensomes for other cellular receptors sheds light on transcripts potentially mediating crosstalk between TRs and these other signaling paradigms. Conclusions: The mouse TR datasets and consensomes are freely available in the Signaling Pathways Project website for hypothesis generation, data validation, and modeling of novel mechanisms of TR regulation of gene expression. Our results demonstrate the insights into the mechanistic basis of thyroid hormone action that can arise from an ongoing commitment on the part of the research community to the deposition of discovery-scale datasets.
Collapse
Affiliation(s)
- Scott A. Ochsner
- The Signaling Pathways Project, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Neil J. McKenna
- The Signaling Pathways Project, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Address correspondence to: Neil J. McKenna, PhD, The Signaling Pathways Project, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
41
|
Kushchayeva YS, Startzell M, Cochran E, Auh S, Sekizkardes H, Soldin SJ, Kushchayev SV, Dieckmann W, Skarulis M, Abdul Sater Z, Brychta RJ, Cypess AM, Lin TC, Lightbourne M, Millo C, Brown RJ. Thyroid Hormone Effects on Glucose Disposal in Patients With Insulin Receptor Mutations. J Clin Endocrinol Metab 2020; 105:5582247. [PMID: 31588494 PMCID: PMC7093053 DOI: 10.1210/clinem/dgz079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/01/2019] [Indexed: 01/16/2023]
Abstract
CONTEXT Patients with mutations of the insulin receptor gene (INSR) have extreme insulin resistance and are at risk for early morbidity and mortality from diabetes complications. A case report suggested that thyroid hormone could improve glycemia in INSR mutation in part by increasing brown adipose tissue (BAT) activity and volume. OBJECTIVE To determine if thyroid hormone increases tissue glucose uptake and improves hyperglycemia in INSR mutation. DESIGN Single-arm, open-label study of liothyronine. SETTING National Institutes of Health. PARTICIPANTS Patients with homozygous (n = 5) or heterozygous (n = 2) INSR mutation. INTERVENTION Liothyronine every 8 hours for 2 weeks (n = 7); additional 6 months' treatment in those with hemoglobin A1c (HbA1c) > 7% (n = 4). OUTCOMES Whole-body glucose uptake by isotopic tracers; tissue glucose uptake in muscle, white adipose tissue (WAT) and BAT by dynamic [18F] fluorodeoxyglucose positron emission tomography/computed tomography; HbA1c. RESULTS There was no change in whole-body, muscle, or WAT glucose uptake from baseline to 2 weeks of liothyronine. After 6 months, there was no change in HbA1c (8.3 ± 1.2 vs 9.1 ± 3.0%, P = 0.27), but there was increased whole-body glucose disposal (22.8 ± 4.9 vs 30.1 ± 10.0 µmol/kg lean body mass/min, P = 0.02), and muscle (0.7 ± 0.1 vs 2.0 ± 0.2 µmol/min/100 mL, P < 0.0001) and WAT glucose uptake (1.2 ± 0.2 vs 2.2 ± 0.3 µmol/min/100 mL, P < 0.0001). BAT glucose uptake could not be quantified because of small volume. There were no signs or symptoms of hyperthyroidism. CONCLUSION Liothyronine administered at well-tolerated doses did not improve HbA1c. However, the observed increases in muscle and WAT glucose uptake support the proposed mechanism that liothyronine increases tissue glucose uptake. More selective agents may be effective at increasing tissue glucose uptake without thyroid hormone-related systemic toxicity.Clinical Trial Registration Number: NCT02457897; https://clinicaltrials.gov/ct2/show/NCT02457897.
Collapse
Affiliation(s)
| | - Megan Startzell
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Elaine Cochran
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Sungyoung Auh
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Hilal Sekizkardes
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Steven J Soldin
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | | | - William Dieckmann
- Positron Emission Tomography Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Monica Skarulis
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Zahraa Abdul Sater
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Robert J Brychta
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Aaron M Cypess
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Tzu-Chun Lin
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Marissa Lightbourne
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Corina Millo
- Positron Emission Tomography Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Rebecca J Brown
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
- Correspondence and Reprint Requests: Rebecca J. Brown, MD, MHSc, Building 10, Room 6-5940, 10 Center Dr., Bethesda, MD 20892. E-mail:
| |
Collapse
|
42
|
Junker D, Syväri J, Weidlich D, Holzapfel C, Drabsch T, Waschulzik B, Rummeny EJ, Hauner H, Karampinos DC. Investigation of the Relationship between MR-Based Supraclavicular Fat Fraction and Thyroid Hormones. Obes Facts 2020; 13:331-343. [PMID: 32564012 PMCID: PMC7445585 DOI: 10.1159/000507294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 03/13/2020] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Brown adipose tissue (BAT) plays a potential role in energy and glucose metabolism in humans. Thyroid hormones (TH) are main regulators of BAT development and function. However, it remains unknown how the magnetic resonance (MR)-based proton density fat fraction (PDFF) of supraclavicular adipose tissue used as a surrogate marker for BAT presence relates to TH. Therefore, the purpose of this analysis was to investigate the relationship between supraclavicular PDFF and serum levels of TH. METHODS In total, 96 adult volunteers from a large cross-sectional study who underwent additional MR examination of the neck and pelvis were included in this analysis. Segmented PDFF maps of the supraclavicular and gluteal subcutaneous adipose tissue were generated. Delta PDFF was calculated as the difference between gluteal and supraclavicular PDFF and grouped as high (≥12%) or low (<12%) based on the median and the clinical rationale of a high versus low probability of BAT being present. Thyroid-stimulating hormone (mIU/L), free triiodothyronine (FT3, pg/mL) and free thyroxine (FT4, ng/dL) levels were determined in blood samples. Body mass index (BMI) was calculated as weight (kg)/height (m)2. Statistical analyses included the use of paired samples ttest, simple linear regression analysis and a multivariable linear regression analysis. RESULTS The median age of the subjects (77% female) was 33 years, BMI ranged from 17.2 to 43.1 kg/m2. Supraclavicular and gluteal PDFF differed significantly (76.5 ± 4.8 vs. 89.4 ± 3.5 %, p < 0.01). Supraclavicular PDFF was associated with FT3 in subjects with high delta PDFF (R2 = 0.17, p < 0.01), with higher FT3 being associated with lower supraclavicular PDFF (y = 85.2 + -3.6 x). In a multivariable linear regression analysis considering further potential prognostic factors, the interaction between the delta PDFF group and FT3 remained a predictor for supraclavicular PDFF (B = -4.65, p < 0.01). DISCUSSION/CONCLUSIONS Supraclavicular PDFF corresponds to the presence of BAT. In the present analysis, supraclavicular PDFF is correlated with FT3 in subjects with high delta PDFF. Therefore, the present findings suggest that biologically active T3 may be involved in the development of supraclavicular BAT.
Collapse
Affiliation(s)
- Daniela Junker
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany,
| | - Jan Syväri
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Dominik Weidlich
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Christina Holzapfel
- Institute for Nutritional Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Theresa Drabsch
- Institute for Nutritional Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Birgit Waschulzik
- Institute of Medical Informatics, Statistics and Epidemiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Ernst J Rummeny
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Hans Hauner
- Institute for Nutritional Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Else Kroener-Fresenius-Center of Nutritional Medicine, ZIEL Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Dimitrios C Karampinos
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
43
|
Abstract
AbstractThyroid hormones are essential for the full thermogenic capacity of brown adipose
tissue. The thermogenic response of brown adipocytes to thyroid hormones is
resulting from the synergistic interaction of thyroid hormones with the
sympathetic nervous system. In recent years, evidence has been provided that
thyroid hormones also induce the browning of white adipose tissues. This review
will provide a brief overview about the recent findings regarding the effects of
thyroid hormones on adipose tissue thermogenesis including central and
peripheral regulation of white adipose tissue browning.
Collapse
Affiliation(s)
- Kerstin Krause
- Department of Endocrinology and Nephrology, University Hospital
Leipzig, Leipzig, Germany
| |
Collapse
|
44
|
Jash S, Banerjee S, Lee MJ, Farmer SR, Puri V. CIDEA Transcriptionally Regulates UCP1 for Britening and Thermogenesis in Human Fat Cells. iScience 2019; 20:73-89. [PMID: 31563853 PMCID: PMC6817690 DOI: 10.1016/j.isci.2019.09.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/02/2019] [Accepted: 09/10/2019] [Indexed: 12/16/2022] Open
Abstract
Our study identifies a transcriptional role of cell death-inducing DNA fragmentation factor-like effector A (CIDEA), a lipid-droplet-associated protein, whereby it regulates human adipocyte britening/beiging with consequences for the regulation of energy expenditure. The comprehensive transcriptome analysis revealed CIDEA's control over thermogenic function in brite/beige human adipocytes. In the absence of CIDEA, achieved by the modified dual-RNA-based CRISPR-Cas9nD10A system, adipocytes lost their britening capability, which was recovered upon CIDEA re-expression. Uncoupling protein 1 (UCP1), the most upregulated gene in brite human adipocytes, was suppressed in CIDEA knockout (KO) primary human adipocytes. Mechanistically, during induced britening, CIDEA shuttled from lipid droplets to the nucleus via an unusual nuclear bipartite signal in a concentration-dependent manner. In the nucleus, it specifically inhibited LXRα repression of UCP1 enhancer activity and strengthened PPARγ binding to UCP1 enhancer, hence driving UCP1 transcription. Overall, our study defines the role of CIDEA in increasing thermogenesis in human adipocytes.
Collapse
Affiliation(s)
- Sukanta Jash
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Sayani Banerjee
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Mi-Jeong Lee
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Stephen R Farmer
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Vishwajeet Puri
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
45
|
Corrêa LH, Heyn GS, Magalhaes KG. The Impact of the Adipose Organ Plasticity on Inflammation and Cancer Progression. Cells 2019; 8:E662. [PMID: 31262098 PMCID: PMC6679170 DOI: 10.3390/cells8070662] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/21/2019] [Accepted: 06/22/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity is characterized by chronic and low-grade systemic inflammation, an increase of adipose tissue, hypertrophy, and hyperplasia of adipocytes. Adipose tissues can be classified into white, brown, beige and pink adipose tissues, which display different regulatory, morphological and functional characteristics of their adipocyte and immune cells. Brown and white adipocytes can play a key role not only in the control of energy homeostasis, or through the balance between energy storage and expenditure, but also by the modulation of immune and inflammatory responses. Therefore, brown and white adipocytes can orchestrate important immunological crosstalk that may deeply impact the tumor microenvironment and be crucial for cancer establishment and progression. Recent works have indicated that white adipose tissues can undergo a process called browning, in which an inducible brown adipocyte develops. In this review, we depict the mechanisms involved in the differential role of brown, white and pink adipocytes, highlighting their structural, morphological, regulatory and functional characteristics and correlation with cancer predisposition, establishment, and progression. We also discuss the impact of the increased adiposity in the inflammatory and immunological modulation. Moreover, we focused on the plasticity of adipocytes, describing the molecules produced and secreted by those cells, the modulation of the signaling pathways involved in the browning phenomena of white adipose tissue and its impact on inflammation and cancer.
Collapse
MESH Headings
- Adipocytes, Brown/immunology
- Adipocytes, Brown/metabolism
- Adipocytes, White/immunology
- Adipocytes, White/metabolism
- Adipose Tissue, Brown/cytology
- Adipose Tissue, Brown/immunology
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/cytology
- Adipose Tissue, White/immunology
- Adipose Tissue, White/metabolism
- Adiposity/immunology
- Animals
- Carcinogenesis/immunology
- Carcinogenesis/pathology
- Disease Models, Animal
- Disease Progression
- Energy Metabolism/immunology
- Humans
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/pathology
- Obesity/complications
- Obesity/immunology
- Obesity/metabolism
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Luís Henrique Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Gabriella Simões Heyn
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Kelly Grace Magalhaes
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia 70910-900, Brazil.
| |
Collapse
|
46
|
Johann K, Cremer AL, Fischer AW, Heine M, Pensado ER, Resch J, Nock S, Virtue S, Harder L, Oelkrug R, Astiz M, Brabant G, Warner A, Vidal-Puig A, Oster H, Boelen A, López M, Heeren J, Dalley JW, Backes H, Mittag J. Thyroid-Hormone-Induced Browning of White Adipose Tissue Does Not Contribute to Thermogenesis and Glucose Consumption. Cell Rep 2019; 27:3385-3400.e3. [DOI: 10.1016/j.celrep.2019.05.054] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 05/07/2019] [Accepted: 05/15/2019] [Indexed: 12/13/2022] Open
|
47
|
Srivastava S, Veech RL. Brown and Brite: The Fat Soldiers in the Anti-obesity Fight. Front Physiol 2019; 10:38. [PMID: 30761017 PMCID: PMC6363669 DOI: 10.3389/fphys.2019.00038] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/14/2019] [Indexed: 12/16/2022] Open
Abstract
Brown adipose tissue (BAT) is proposed to maintain thermal homeostasis through dissipation of chemical energy as heat by the uncoupling proteins (UCPs) present in their mitochondria. The recent demonstration of the presence of BAT in humans has invigorated research in this area. The research has provided many new insights into the biology and functioning of this tissue and the biological implications of its altered activities. Another finding of interest is browning of white adipose tissue (WAT) resulting in what is known as beige/brite cells, which have increased mitochondrial proteins and UCPs. In general, it has been observed that the activation of BAT is associated with various physiological improvements such as a reduction in blood glucose levels increased resting energy expenditure and reduced weight. Given the similar physiological functions of BAT and beige/ brite cells and the higher mass of WAT compared to BAT, it is likely that increasing the brite/beige cells in WATs may also lead to greater metabolic benefits. However, development of treatments targeting brown fat or WAT browning would require not only a substantial understanding of the biology of these tissues but also the effect of altering their activity levels on whole body metabolism and physiology. In this review, we present evidence from recent literature on the substrates utilized by BAT, regulation of BAT activity and browning by circulating molecules. We also present dietary and pharmacological activators of brown and beige/brite adipose tissue and the effect of physical exercise on BAT activity and browning.
Collapse
Affiliation(s)
- Shireesh Srivastava
- Systems Biology for Biofuels Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Richard L Veech
- Laboratory of Metabolic Control, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
48
|
Yau WW, Singh BK, Lesmana R, Zhou J, Sinha RA, Wong KA, Wu Y, Bay BH, Sugii S, Sun L, Yen PM. Thyroid hormone (T 3) stimulates brown adipose tissue activation via mitochondrial biogenesis and MTOR-mediated mitophagy. Autophagy 2019; 15:131-150. [PMID: 30209975 PMCID: PMC6287687 DOI: 10.1080/15548627.2018.1511263] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/26/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022] Open
Abstract
The thyroid hormone triiodothyronine (T3) activates thermogenesis by uncoupling electron transport from ATP synthesis in brown adipose tissue (BAT) mitochondria. Although T3 can induce thermogenesis by sympathetic innervation, little is known about its cell autonomous effects on BAT mitochondria. We thus examined effects of T3 on mitochondrial activity, autophagy, and metabolism in primary brown adipocytes and BAT and found that T3 increased fatty acid oxidation and mitochondrial respiration as well as autophagic flux, mitophagy, and mitochondrial biogenesis. Interestingly, there was no significant induction of intracellular reactive oxygen species (ROS) despite high mitochondrial respiration and UCP1 induction by T3. However, when cells were treated with Atg5 siRNA to block autophagy, induction of mitochondrial respiration by T3 decreased, and was accompanied by ROS accumulation, demonstrating a critical role for autophagic mitochondrial turnover. We next generated an Atg5 conditional knockout mouse model (Atg5 cKO) by injecting Ucp1 promoter-driven Cre-expressing adenovirus into Atg5Flox/Flox mice to examine effects of BAT-specific autophagy on thermogenesis in vivo. Hyperthyroid Atg5 cKO mice exhibited lower body temperature than hyperthyroid or euthyroid control mice. Metabolomic analysis showed that T3 increased short and long chain acylcarnitines in BAT, consistent with increased β-oxidation. T3 also decreased amino acid levels, and in conjunction with SIRT1 activation, decreased MTOR activity to stimulate autophagy. In summary, T3 has direct effects on mitochondrial autophagy, activity, and turnover in BAT that are essential for thermogenesis. Stimulation of BAT activity by thyroid hormone or its analogs may represent a potential therapeutic strategy for obesity and metabolic diseases. Abbreviations: ACACA: acetyl-Coenzyme A carboxylase alpha; AMPK: AMP-activated protein kinase; Acsl1: acyl-CoA synthetase long-chain family member 1; ATG5: autophagy related 5; ATG7: autophagy related 7; ATP: adenosine triphosphate; BAT: brown adipose tissue; cKO: conditional knockout; COX4I1: cytochrome c oxidase subunit 4I1; Cpt1b: carnitine palmitoyltransferase 1b, muscle; CQ: chloroquine; DAPI: 4',6-diamidino-2-phenylindole; DIO2: deiodinase, iodothyronine, type 2; DMEM: Dulbecco's modified Eagle's medium; EIF4EBP1: eukaryotic translation initiation factor 4E binding protein 1; Fabp4: fatty acid binding protein 4, adipocyte; FBS: fetal bovine serum; FCCP: carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone; FGF: fibroblast growth factor; FOXO1: forkhead box O1; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; Gpx1: glutathione peroxidase 1; Lipe: lipase, hormone sensitive; MAP1LC3B: microtubule-associated protein 1 light chain 3; mRNA: messenger RNA; MTORC1: mechanistic target of rapamycin kinase complex 1; NAD: nicotinamide adenine dinucleotide; Nrf1: nuclear respiratory factor 1; OCR: oxygen consumption rate; PBS: phosphate-buffered saline; PCR: polymerase chain reaction; PPARGC1A: peroxisome proliferative activated receptor, gamma, coactivator 1 alpha; Pnpla2: patatin-like phospholipase domain containing 2; Prdm16: PR domain containing 16; PRKA: protein kinase, AMP-activated; RPS6KB: ribosomal protein S6 kinase; RFP: red fluorescent protein; ROS: reactive oxygen species; SD: standard deviation; SEM: standard error of the mean; siRNA: small interfering RNA; SIRT1: sirtuin 1; Sod1: superoxide dismutase 1, soluble; Sod2: superoxide dismutase 2, mitochondrial; SQSTM1: sequestosome 1; T3: 3,5,3'-triiodothyronine; TFEB: transcription factor EB; TOMM20: translocase of outer mitochondrial membrane 20; UCP1: uncoupling protein 1 (mitochondrial, proton carrier); ULK1: unc-51 like kinase 1; VDAC1: voltage-dependent anion channel 1; WAT: white adipose tissue.
Collapse
Affiliation(s)
- Winifred W. Yau
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Brijesh K. Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Ronny Lesmana
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
- Physiology Division, Department of Anatomy, Physiology and Biology Cell, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Central laboratory, Universitas Padjadjaran, Bandung, Indonesia
| | - Jin Zhou
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Rohit A. Sinha
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Kiraely A. Wong
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Yajun Wu
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shigeki Sugii
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
- Fat Metabolism and Stem Cell Group, Singapore Bioimaging Consortium, A*STAR, Singapore
| | - Lei Sun
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Paul M. Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
- Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
49
|
Kaisanlahti A, Glumoff T. Browning of white fat: agents and implications for beige adipose tissue to type 2 diabetes. J Physiol Biochem 2018; 75:1-10. [PMID: 30506389 PMCID: PMC6513802 DOI: 10.1007/s13105-018-0658-5] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/31/2018] [Indexed: 12/23/2022]
Abstract
Mammalian adipose tissue is traditionally categorized into white and brown relating to their function and morphology: while white serves as an energy storage, brown adipose tissue acts as the heat generator maintaining the core body temperature. The most recently identified type of fat, beige adipocyte tissue, resembles brown fat by morphology and function but is developmentally more related to white. The synthesis of beige fat, so-called browning of white fat, has developed into a topical issue in diabetes and metabolism research. This is due to its favorable effect on whole-body energy metabolism and the fact that it can be recruited during adult life. Indeed, brown and beige adipose tissues have been demonstrated to play a role in glucose homeostasis, insulin sensitivity, and lipid metabolism—all factors related to pathogenesis of type 2 diabetes. Many agents capable of initiating browning have been identified so far and tested widely in humans and animal models including in vitro and in vivo experiments. Interestingly, several agents demonstrated to have browning activity are in fact secreted as adipokines from brown and beige fat tissue, suggesting a physiological relevance both in beige adipocyte recruitment processes and in maintenance of metabolic homeostasis. The newest findings on agents driving beige fat recruitment, their mechanisms, and implications on type 2 diabetes are discussed in this review.
Collapse
MESH Headings
- Adipose Tissue, Beige/drug effects
- Adipose Tissue, Beige/metabolism
- Adipose Tissue, Beige/pathology
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/pathology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Animals
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Energy Metabolism/drug effects
- Energy Metabolism/genetics
- Glucagon-Like Peptide 1/pharmacology
- Glucose/metabolism
- Humans
- Insulin Resistance
- Leptin/pharmacology
- Lipid Metabolism/drug effects
- Lipid Metabolism/genetics
- Lipotropic Agents/pharmacology
- Melatonin/pharmacology
- Natriuretic Peptides/pharmacology
- Thermogenesis/drug effects
- Thermogenesis/genetics
- Tretinoin/pharmacology
Collapse
Affiliation(s)
- A Kaisanlahti
- Biocenter Oulu/Cancer Research and Translational Medicine Research Unit, University of Oulu, Aapistie 5, P.O. Box 5281, 90014, Oulu, Finland.
| | - T Glumoff
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7A, P.O Box 5400, 90014, Oulu, Finland
| |
Collapse
|
50
|
Zhang Z, Boelen A, Kalsbeek A, Fliers E. TRH Neurons and Thyroid Hormone Coordinate the Hypothalamic Response to Cold. Eur Thyroid J 2018; 7:279-288. [PMID: 30574457 PMCID: PMC6276749 DOI: 10.1159/000493976] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/21/2018] [Indexed: 12/28/2022] Open
Abstract
Thyroid hormone (TH) plays a key role in regulating body temperature in mammals. Cold exposure stimulates the hypothalamus-pituitary-thyroid (HPT) axis at the hypothalamic level by activating hypophysiotropic thyrotropin-releasing hormone (TRH)-producing neurons, ultimately resulting in increased plasma TH concentrations. Importantly, the local TH metabolism within various cold-responsive organs enables tissue-specific action of TH on heat production and adaption to cold independently of the circulating TH levels. In addition to these neuroendocrine effects, TRH neurons in the hypothalamus also have neural connections with brown adipose tissue (BAT), probably contributing to regulation of thermogenesis by the autonomic nervous system. Recent studies have demonstrated that intrahypothalamic TH has profound metabolic effects on BAT, the liver, and the heart that are mediated via the autonomic nervous system. These effects originate in various hypothalamic nuclei, including the paraventricular nucleus (PVN), the ventromedial nucleus, and recently reported neurons in the anterior hypothalamic area, indicating a potential central function for TH on thermoregulation. Finally, although robust stimulation of the thermogenic program in BAT was shown upon TH administration in the ventromedial hypothalamus, the physiological relevance of these neurally mediated effects of TH is unclear at present. This review provides an overview of studies reporting the role of TH in cold defense, with a focus on recent literature evidencing the centrally mediated effects of TRH and TH.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anita Boelen
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- *Eric Fliers, Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, NL–1105AZ Amsterdam (The Netherlands), E-Mail
| |
Collapse
|