1
|
Zhu H, Xu H, Zhang Y, Brodský J, Gablech I, Korabečná M, Neuzil P. Exploring the Frontiers of Cell Temperature Measurement and Thermogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2402135. [PMID: 39467049 DOI: 10.1002/advs.202402135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/24/2024] [Indexed: 10/30/2024]
Abstract
The precise measurement of cell temperature and an in-depth understanding of thermogenic processes are critical in unraveling the complexities of cellular metabolism and its implications for health and disease. This review focuses on the mechanisms of local temperature generation within cells and the array of methods developed for accurate temperature assessment. The contact and noncontact techniques are introduced, including infrared thermography, fluorescence thermometry, and other innovative approaches to localized temperature measurement. The role of thermogenesis in cellular metabolism, highlighting the integral function of temperature regulation in cellular processes, environmental adaptation, and the implications of thermogenic dysregulation in diseases such as metabolic disorders and cancer are further discussed. The challenges and limitations in this field are critically analyzed while technological advancements and future directions are proposed to overcome these barriers. This review aims to provide a consolidated resource for current methodologies, stimulate discussion on the limitations and challenges, and inspire future innovations in the study of cellular thermodynamics.
Collapse
Affiliation(s)
- Hanliang Zhu
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, P. R. China
| | - Haotian Xu
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, P. R. China
| | - Yue Zhang
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, P. R. China
| | - Jan Brodský
- Department of Microelectronics, The Faculty of Electrical Engineering and Communication Technology, Brno University of Technology, Technická 3058/10, Brno, 616 00, Czech Republic
| | - Imrich Gablech
- Department of Microelectronics, The Faculty of Electrical Engineering and Communication Technology, Brno University of Technology, Technická 3058/10, Brno, 616 00, Czech Republic
| | - Marie Korabečná
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Albertov 4, Prague, 128 00, Czech Republic
- Department of Laboratory Medicine, Faculty of Health Care and Social Work, University of Trnava in Trnava, Universitne namestie 1, Trnava, 918 43, Slovakia
| | - Pavel Neuzil
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, P. R. China
| |
Collapse
|
2
|
Abobeleira JP, Neto AC, Mauersberger J, Salazar M, Botelho M, Fernandes AS, Martinho M, Serrão MP, Rodrigues AR, Almeida H, Gouveia AM, Neves D. Evidence of Browning and Inflammation Features in Visceral Adipose Tissue of Women with Endometriosis. Arch Med Res 2024; 55:103064. [PMID: 39244839 DOI: 10.1016/j.arcmed.2024.103064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/28/2024] [Accepted: 07/24/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Patients with endometriosis tend to have a low body mass index, suggesting an inverse relationship between body fat and risk of disease. This is supported by evidence that miRNAs differentially expressed in endometriosis induce browning of pre-adipocytes in vitro. Thus, we hypothesize that endometriosis may underlie adipose tissue (AT) dysfunction and browning. AIMS Identify inflammation and browning processes in AT collected from endometriosis patients. METHODS Visceral and subcutaneous AT samples were obtained during endometriosis (n = 32) or uterine myoma (n = 14; controls) surgery. Blood catecholamines were determined by high-performance liquid chromatography while IL-6 and TGF-β levels were quantified by ELISA. Adipocyte cross-sectional areas were analyzed in H&E-stained sections by computer-assisted morphometry. Macrophages (F4/80; Galectin-3) and browning activation (UCP-1; PGC-1α) in tissues were identified by dual label immunofluorescence. Expression of inflammatory (IL-6; MCP-1; Galectin-3; CD206; TIMP1; TGF-β) and browning-related (UCP-1; PGC-1α; DIO2; CITED1; CIDEA; TMEM26; TBX1; PRDM16; PPAR-γ) molecules in AT were assessed by RT-PCR and Western blotting. RESULTS Compared to controls, patients presented smaller adipocytes, especially in VAT, and lower norepinephrine levels. Serum IL-6, but not TGF-β, was increased in patients. UCP-1, PGC-1α, IL-6, and MCP-1 were upregulated in VAT from endometriosis women, which also evidenced a reduction of CD206, relative to controls. However, no differences were found in mRNA expression of IL-6, TIMP1, and TGF-β nor Galectin-3 protein levels. In SAT, protein expression remained unchanged between patients and controls. CONCLUSIONS Our findings support an endometriosis' role as a pro-catabolic state along with local signals of VAT browning and inflammation.
Collapse
Affiliation(s)
- José Pedro Abobeleira
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde, i3S, Porto, Portugal
| | - Ana Catarina Neto
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde, i3S, Porto, Portugal
| | - Jan Mauersberger
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde, i3S, Porto, Portugal
| | - Maria Salazar
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde, i3S, Porto, Portugal
| | - Maria Botelho
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde, i3S, Porto, Portugal
| | - Ana Sofia Fernandes
- Department of Obstetrics and Gynecology, Centro Hospitalar Universitário S. João, Porto, Portugal
| | - Margarida Martinho
- Department of Obstetrics and Gynecology, Centro Hospitalar Universitário S. João, Porto, Portugal
| | - Maria Paula Serrão
- Department of Biomedicine-Pharmacology and Therapeutics Unit, Faculty of Medicine of the University of Porto, Porto, Portugal; MedInUP, Center for Drug Discovery and Innovative Medicines, Porto, Portugal
| | - Adriana Raquel Rodrigues
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde, i3S, Porto, Portugal; Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
| | - Henrique Almeida
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde, i3S, Porto, Portugal
| | - Alexandra Maria Gouveia
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde, i3S, Porto, Portugal
| | - Delminda Neves
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde, i3S, Porto, Portugal.
| |
Collapse
|
3
|
Ishida Y, Matsushita M, Yoneshiro T, Saito M, Fuse S, Hamaoka T, Kuroiwa M, Tanaka R, Kurosawa Y, Nishimura T, Motoi M, Maeda T, Nakayama K. Genetic evidence for involvement of β2-adrenergic receptor in brown adipose tissue thermogenesis in humans. Int J Obes (Lond) 2024; 48:1110-1117. [PMID: 38632325 PMCID: PMC11281906 DOI: 10.1038/s41366-024-01522-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Sympathetic activation of brown adipose tissue (BAT) thermogenesis can ameliorate obesity and related metabolic abnormalities. However, crucial subtypes of the β-adrenergic receptor (AR), as well as effects of its genetic variants on functions of BAT, remains unclear in humans. We conducted association analyses of genes encoding β-ARs and BAT activity in human adults. METHODS Single nucleotide polymorphisms (SNPs) in β1-, β2-, and β3-AR genes (ADRB1, ADRB2, and ADRB3) were tested for the association with BAT activity under mild cold exposure (19 °C, 2 h) in 399 healthy Japanese adults. BAT activity was measured using fluorodeoxyglucose-positron emission tomography and computed tomography (FDG-PET/CT). To validate the results, we assessed the effects of SNPs in the two independent populations comprising 277 healthy East Asian adults using near-infrared time-resolved spectroscopy (NIRTRS) or infrared thermography (IRT). Effects of SNPs on physiological responses to intensive cold exposure were tested in 42 healthy Japanese adult males using an artificial climate chamber. RESULTS We found a significant association between a functional SNP (rs1042718) in ADRB2 and BAT activity assessed with FDG-PET/CT (p < 0.001). This SNP also showed an association with cold-induced thermogenesis in the population subset. Furthermore, the association was replicated in the two other independent populations; BAT activity was evaluated by NIRTRS or IRT (p < 0.05). This SNP did not show associations with oxygen consumption and cold-induced thermogenesis under intensive cold exposure, suggesting the irrelevance of shivering thermogenesis. The SNPs of ADRB1 and ADRB3 were not associated with these BAT-related traits. CONCLUSIONS The present study supports the importance of β2-AR in the sympathetic regulation of BAT thermogenesis in humans. The present collection of DNA samples is the largest to which information on the donor's BAT activity has been assigned and can serve as a reference for further in-depth understanding of human BAT function.
Collapse
MESH Headings
- Humans
- Thermogenesis/physiology
- Thermogenesis/genetics
- Adipose Tissue, Brown/metabolism
- Male
- Adult
- Polymorphism, Single Nucleotide
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Female
- Middle Aged
- Japan
- Positron Emission Tomography Computed Tomography
- Receptors, Adrenergic, beta-3/genetics
- Receptors, Adrenergic, beta-3/metabolism
- Asian People/genetics
Collapse
Affiliation(s)
- Yuka Ishida
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, 277-8562, Japan
| | - Mami Matsushita
- Department of Nutrition, School of Nursing and Nutrition, Tenshi College, Sapporo, Hokkaido, 065-0013, Japan
| | - Takeshi Yoneshiro
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo, 153-8904, Japan
- Department of Molecular Metabolism and Physiology, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, 980-8575, Japan
| | - Masayuki Saito
- Department of Nutrition, School of Nursing and Nutrition, Tenshi College, Sapporo, Hokkaido, 065-0013, Japan
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Sayuri Fuse
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Takafumi Hamaoka
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Miyuki Kuroiwa
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Riki Tanaka
- Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Fukuoka, 814-0180, Japan
| | - Yuko Kurosawa
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Takayuki Nishimura
- Department of Human Life Design and Science, Faculty of Design, Kyushu University, Fukuoka, Fukuoka, 815-8540, Japan
- Physiological Anthropology Research Center, Faculty of Design, Kyushu University, Fukuoka, Fukuoka, 815-8540, Japan
| | - Midori Motoi
- Department of Human Life Design and Science, Faculty of Design, Kyushu University, Fukuoka, Fukuoka, 815-8540, Japan
| | - Takafumi Maeda
- Department of Human Life Design and Science, Faculty of Design, Kyushu University, Fukuoka, Fukuoka, 815-8540, Japan
- Physiological Anthropology Research Center, Faculty of Design, Kyushu University, Fukuoka, Fukuoka, 815-8540, Japan
| | - Kazuhiro Nakayama
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, 277-8562, Japan.
| |
Collapse
|
4
|
Chen X, Wang Y, Li H, Deng Y, Giang C, Song A, Liu Y, Wang QA, Zhu Y. Hyaluronan Mediates Cold-Induced Adipose Tissue Beiging. Cells 2024; 13:1233. [PMID: 39120264 PMCID: PMC11311271 DOI: 10.3390/cells13151233] [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: 05/30/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Adipose tissue beiging refers to the process by which beige adipocytes emerge in classical white adipose tissue depots. Beige adipocytes dissipate chemical energy and secrete adipokines, such as classical brown adipocytes, to improve systemic metabolism, which is beneficial for people with obesity and metabolic diseases. Cold exposure and β3-adrenergic receptor (AR) agonist treatment are two commonly used stimuli for increasing beige adipocytes in mice; however, their underlying biological processes are different. Transcriptional analysis of inguinal white adipose tissue (iWAT) has revealed that changes in extracellular matrix (ECM) pathway genes are specific to cold exposure. Hyaluronic acid (HA), a non-sulfated linear polysaccharide produced by nearly all cells, is one of the most common components of ECM. We found that cold exposure significantly increased iWAT HA levels, whereas the β3-AR agonist CL316,243 did not. Increasing HA levels in iWAT by Has2 overexpression significantly increases cold-induced adipose tissue beiging; in contrast, decreasing HA by Spam1 overexpression, which encodes a hyaluronidase that digests HA, significantly decreases cold-induced iWAT beiging. All these data implicate a role of HA in promoting adipose tissue beiging, which is unique to cold exposure. Given the failure of β3-AR agonists in clinical trials for obesity and metabolic diseases, increasing HA could serve as a new approach for recruiting more beige adipocytes to combat metabolic diseases.
Collapse
Affiliation(s)
- Xi Chen
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yifan Wang
- Department of Molecular Endocrinology, Diabetes and Metabolism Institute, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Huiqiao Li
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yanru Deng
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Charlise Giang
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anying Song
- Department of Molecular Endocrinology, Diabetes and Metabolism Institute, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Yu’e Liu
- Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai 200092, China
| | - Qiong A. Wang
- Department of Molecular Endocrinology, Diabetes and Metabolism Institute, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Yi Zhu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Prapaharan B, Lea M, Beaudry JL. Weighing in on the role of brown adipose tissue for treatment of obesity. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13157. [PMID: 39087083 PMCID: PMC11290130 DOI: 10.3389/jpps.2024.13157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024]
Abstract
Brown adipose tissue (BAT) activation is an emerging target for obesity treatments due to its thermogenic properties stemming from its ability to shuttle energy through uncoupling protein 1 (Ucp1). Recent rodent studies show how BAT and white adipose tissue (WAT) activity can be modulated to increase the expression of thermogenic proteins. Consequently, these alterations enable organisms to endure cold-temperatures and elevate energy expenditure, thereby promoting weight loss. In humans, BAT is less abundant in obese subjects and impacts of thermogenesis are less pronounced, bringing into question whether energy expending properties of BAT seen in rodents can be translated to human models. Our review will discuss pharmacological, hormonal, bioactive, sex-specific and environmental activators and inhibitors of BAT to determine the potential for BAT to act as a therapeutic strategy. We aim to address the feasibility of utilizing BAT modulators for weight reduction in obese individuals, as recent studies suggest that BAT's contributions to energy expenditure along with Ucp1-dependent and -independent pathways may or may not rectify energy imbalance characteristic of obesity.
Collapse
Affiliation(s)
| | | | - Jacqueline L. Beaudry
- Temerty Faculty of Medicine, Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
6
|
Li L, Feldman BJ. White adipocytes in subcutaneous fat depots require KLF15 for maintenance in preclinical models. J Clin Invest 2024; 134:e172360. [PMID: 38949025 PMCID: PMC11213504 DOI: 10.1172/jci172360] [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: 05/18/2023] [Accepted: 05/10/2024] [Indexed: 07/02/2024] Open
Abstract
Healthy adipose tissue is essential for normal physiology. There are 2 broad types of adipose tissue depots: brown adipose tissue (BAT), which contains adipocytes poised to burn energy through thermogenesis, and white adipose tissue (WAT), which contains adipocytes that store lipids. However, within those types of adipose, adipocytes possess depot and cell-specific properties that have important implications. For example, the subcutaneous and visceral WAT confers divergent risk for metabolic disease. Further, within a depot, different adipocytes can have distinct properties; subcutaneous WAT can contain adipocytes with either white or brown-like (beige) adipocyte properties. However, the pathways that regulate and maintain this cell and depot-specificity are incompletely understood. Here, we found that the transcription factor KLF15 is required for maintaining white adipocyte properties selectively within the subcutaneous WAT. We revealed that deletion of Klf15 is sufficient to induce beige adipocyte properties and that KLF15's direct regulation of Adrb1 is a critical molecular mechanism for this process. We uncovered that this activity is cell autonomous but has systemic implications in mouse models and is conserved in primary human adipose cells. Our results elucidate a pathway for depot-specific maintenance of white adipocyte properties that could enable the development of therapies for obesity and associated diseases.
Collapse
Affiliation(s)
- Liang Li
- Department of Pediatrics, University of California, San Francisco (UCSF) School of Medicine, San Francisco, California, USA
| | - Brian J. Feldman
- Department of Pediatrics, University of California, San Francisco (UCSF) School of Medicine, San Francisco, California, USA
- Nutrition and Obesity Research Center, UCSF, San Francisco, California, USA
| |
Collapse
|
7
|
Dumont L, Caron A, Richard G, Croteau E, Fortin M, Frisch F, Phoenix S, Dubreuil S, Guérin B, Turcotte ÉE, Carpentier AC, Blondin DP. The effects of the β 1-adrenergic receptor antagonist bisoprolol administration on mirabegron-stimulated human brown adipose tissue thermogenesis. Acta Physiol (Oxf) 2024; 240:e14127. [PMID: 38502056 DOI: 10.1111/apha.14127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 01/25/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024]
Abstract
AIM Pharmacological stimulation of human brown adipose tissue (BAT) has been hindered by ineffective activation or undesirable off-target effects. Oral administration of the maximal allowable dose of mirabegron (200 mg), a β3-adrenergic receptor (β3-AR) agonist, has been effective in stimulating BAT thermogenesis and whole-body energy expenditure. However, this has been accompanied by undesirable cardiovascular effects. Therefore, we hypothesized that combining mirabegron with a β1-AR antagonist could suppress these unwanted effects and increase the stimulation of the β3-AR and β2-AR in BAT. METHODS We performed a randomized crossover trial (NCT04823442) in 8 lean men. Mirabegron (200 mg) was administered orally with or without the β1-AR antagonist bisoprolol (10 mg). Dynamic [11C]-acetate and 2-deoxy-2-[18F]fluoro-d-glucose PET/CT scans were performed sequentially after oral administration of mirabegron ± bisoprolol. RESULTS Compared to room temperature, mirabegron alone increased BAT oxidative metabolism (0.84 ± 0.46 vs. 1.79 ± 0.91 min-1, p = 0.0433), but not when combined with bisoprolol. The metabolic rate of glucose in BAT, measured using [18F]FDG PET, was significantly higher with mirabegron than mirabegron with bisoprolol (24 ± 10 vs. 16 ± 8 nmol/g/min, p = 0.0284). Bisoprolol inhibited the mirabegron-induced increase in systolic blood pressure and heart rate. CONCLUSION The administration of bisoprolol decreases the adverse cardiovascular effects of mirabegron. However, the provided dose also blunted the mirabegron-stimulated increase in BAT lipolysis, thermogenesis, and glucose uptake. The attenuation in BAT blood flow induced by the large dose of bisoprolol may have limited BAT thermogenesis.
Collapse
Affiliation(s)
- Lauralyne Dumont
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
- Department of Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Quebec City, Québec, Canada
- Quebec Heart and Lung Institute, Quebec City, Québec, Canada
- Montreal Diabetes Research Center, Montreal, Québec, Canada
| | - Gabriel Richard
- Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Etienne Croteau
- Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Mélanie Fortin
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Frédérique Frisch
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Serge Phoenix
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
- Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Stéphanie Dubreuil
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Brigitte Guérin
- Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Éric E Turcotte
- Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - André C Carpentier
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Denis P Blondin
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
8
|
Gómez-Hernández A, de las Heras N, Gálvez BG, Fernández-Marcelo T, Fernández-Millán E, Escribano Ó. New Mediators in the Crosstalk between Different Adipose Tissues. Int J Mol Sci 2024; 25:4659. [PMID: 38731880 PMCID: PMC11083914 DOI: 10.3390/ijms25094659] [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: 03/22/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Adipose tissue is a multifunctional organ that regulates many physiological processes such as energy homeostasis, nutrition, the regulation of insulin sensitivity, body temperature, and immune response. In this review, we highlight the relevance of the different mediators that control adipose tissue activity through a systematic review of the main players present in white and brown adipose tissues. Among them, inflammatory mediators secreted by the adipose tissue, such as classical adipokines and more recent ones, elements of the immune system infiltrated into the adipose tissue (certain cell types and interleukins), as well as the role of intestinal microbiota and derived metabolites, have been reviewed. Furthermore, anti-obesity mediators that promote the activation of beige adipose tissue, e.g., myokines, thyroid hormones, amino acids, and both long and micro RNAs, are exhaustively examined. Finally, we also analyze therapeutic strategies based on those mediators that have been described to date. In conclusion, novel regulators of obesity, such as microRNAs or microbiota, are being characterized and are promising tools to treat obesity in the future.
Collapse
Affiliation(s)
- Almudena Gómez-Hernández
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain; (A.G.-H.); (B.G.G.); (T.F.-M.); (E.F.-M.)
| | - Natalia de las Heras
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain;
| | - Beatriz G. Gálvez
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain; (A.G.-H.); (B.G.G.); (T.F.-M.); (E.F.-M.)
| | - Tamara Fernández-Marcelo
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain; (A.G.-H.); (B.G.G.); (T.F.-M.); (E.F.-M.)
| | - Elisa Fernández-Millán
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain; (A.G.-H.); (B.G.G.); (T.F.-M.); (E.F.-M.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Óscar Escribano
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain; (A.G.-H.); (B.G.G.); (T.F.-M.); (E.F.-M.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
9
|
Kim S, Yazawa T, Koide A, Yoneda E, Aoki R, Okazaki T, Tomita K, Watanabe H, Muroi Y, Testuka M, Muranishi Y. Potential Role of Pig UCP3 in Modulating Adipocyte Browning via the Beta-Adrenergic Receptor Signaling Pathway. BIOLOGY 2024; 13:284. [PMID: 38785767 PMCID: PMC11117546 DOI: 10.3390/biology13050284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024]
Abstract
Adipose tissue plays an important role in regulating body temperature and metabolism, with white adipocytes serving as storage units for energy. Recent research focused on the browning of white adipocytes (beige adipocytes), causing thermogenesis and lipolysis. The process of browning is linked to the activation of uncoupling protein (UCP) expression, which can be mediated by the β3 adrenergic receptor pathway. Transcriptional factors, such as peroxisome proliferator activated receptor γ (PPARγ) and PPARγ coactivator 1 alpha, play vital roles in cell fate determination for fat cells. Beige adipocytes have metabolic therapeutic potential to combat diseases such as obesity, diabetes mellitus, and dyslipidemia, owing to their significant impact on metabolic functions. However, the molecular mechanisms that cause the induction of browning are unclear. Therefore, research using animal models and primary culture is essential to provide an understanding of browning for further application in human metabolic studies. Pigs have physiological similarities to humans; hence, they are valuable models for research on adipose tissue. This study demonstrates the browning potential of pig white adipocytes through primary culture experiments. The results show that upregulation of UCP3 gene expression and fragmentation of lipid droplets into smaller particles occur due to isoproterenol stimulation, which activates beta-adrenergic receptor signaling. Furthermore, PPARγ and PGC-1α were found to activate the UCP3 promoter region, similar to that of UCP1. These findings suggest that pigs undergo metabolic changes that induce browning in white adipocytes, providing a promising approach for metabolic research with potential implications for human health. This study offers valuable insights into the mechanism of adipocyte browning using pig primary culture that can enhance our understanding of human metabolism, leading to cures for commonly occurring diseases.
Collapse
Affiliation(s)
- Sangwoo Kim
- School of Agriculture and Animal Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Hokkaido, Japan (E.Y.); (R.A.); (T.O.); (K.T.); (H.W.); (Y.M.); (M.T.)
| | - Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, Asahikawa 078-8510, Hokkaido, Japan;
| | - Akari Koide
- School of Agriculture and Animal Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Hokkaido, Japan (E.Y.); (R.A.); (T.O.); (K.T.); (H.W.); (Y.M.); (M.T.)
| | - Erina Yoneda
- School of Agriculture and Animal Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Hokkaido, Japan (E.Y.); (R.A.); (T.O.); (K.T.); (H.W.); (Y.M.); (M.T.)
| | - Risa Aoki
- School of Agriculture and Animal Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Hokkaido, Japan (E.Y.); (R.A.); (T.O.); (K.T.); (H.W.); (Y.M.); (M.T.)
| | - Tatsuki Okazaki
- School of Agriculture and Animal Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Hokkaido, Japan (E.Y.); (R.A.); (T.O.); (K.T.); (H.W.); (Y.M.); (M.T.)
| | - Kisaki Tomita
- School of Agriculture and Animal Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Hokkaido, Japan (E.Y.); (R.A.); (T.O.); (K.T.); (H.W.); (Y.M.); (M.T.)
| | - Hiroyuki Watanabe
- School of Agriculture and Animal Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Hokkaido, Japan (E.Y.); (R.A.); (T.O.); (K.T.); (H.W.); (Y.M.); (M.T.)
| | - Yoshikage Muroi
- School of Agriculture and Animal Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Hokkaido, Japan (E.Y.); (R.A.); (T.O.); (K.T.); (H.W.); (Y.M.); (M.T.)
| | - Masafumi Testuka
- School of Agriculture and Animal Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Hokkaido, Japan (E.Y.); (R.A.); (T.O.); (K.T.); (H.W.); (Y.M.); (M.T.)
| | - Yuki Muranishi
- School of Agriculture and Animal Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Hokkaido, Japan (E.Y.); (R.A.); (T.O.); (K.T.); (H.W.); (Y.M.); (M.T.)
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Suita 565-0871, Osaka, Japan
| |
Collapse
|
10
|
Yang S, Ma H, Wang L, Wang F, Xia J, Liu D, Mu L, Yang X, Liu D. The Role of β3-Adrenergic Receptors in Cold-Induced Beige Adipocyte Production in Pigs. Cells 2024; 13:709. [PMID: 38667324 PMCID: PMC11049327 DOI: 10.3390/cells13080709] [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: 02/29/2024] [Revised: 04/13/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
After exposure to cold stress, animals enhance the production of beige adipocytes and expedite thermogenesis, leading to improved metabolic health. Although brown adipose tissue in rodents is primarily induced by β3-adrenergic receptor (ADRB3) stimulation, the activation of major β-adrenergic receptors (ADRBs) in pigs has been a topic of debate. To address this, we developed overexpression vectors for ADRB1, ADRB2, and ADRB3 and silenced the expression of these receptors to observe their effects on the adipogenic differentiation stages of porcine preadipocytes. Our investigation revealed that cold stress triggers the transformation of subcutaneous white adipose tissue to beige adipose tissue in pigs by modulating adrenergic receptor levels. Meanwhile, we found that ADRB3 promotes the transformation of white adipocytes into beige adipocytes. Notably, ADRB3 enhances the expression of beige adipose tissue marker genes, consequently influencing cellular respiration and metabolism by regulating lipolysis and mitochondrial expression. Therefore, ADRB3 may serve as a pivotal gene in animal husbandry and contribute to the improvement of cold intolerance in piglets.
Collapse
Affiliation(s)
- Shuo Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Hong Ma
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Liang Wang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Fang Wang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Jiqiao Xia
- College of Animal Science and Technology, Hebei Normal University of Science & Technology, Qinhuangdao 066004, China
| | - Dongyu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Linlin Mu
- Institute of Forage and Grassland Sciences, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Xiuqin Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Di Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| |
Collapse
|
11
|
Soriano-Ursúa MA, Arias-Montaño JA, Ocampo-Néstor AL, Hernández-Martínez CF, Santillán-Torres I, Andrade-Jorge E, Valdez-Ortiz R, Fernández-Del Valle C, Trujillo-Ferrara JG. In silico identification of a biarylamine acting as agonist at human β 3 adrenoceptors and exerting BRL37344-like effects on mouse metabolism. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2159-2170. [PMID: 37792048 DOI: 10.1007/s00210-023-02753-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/26/2023] [Indexed: 10/05/2023]
Abstract
Human β3-adrenoceptor (β3AR) agonists were considered potential agents for the treatment of metabolic disorders. However, compounds tested as β3AR ligands have shown marked differences in pharmacological profile in rodent and human species, although these compounds remain attractive as they were successfully repurposed for the therapy of urinary incontinence. In this work, some biarylamine compounds were designed and tested in silico as potential β3AR agonists on 3-D models of mouse or human β3ARs. Based on the theoretical results, we identified, synthesized and tested a biarylamine compound (polibegron). In CHO-K1 cells expressing the human β3AR, polibegron and the β3AR agonist BRL 37344 were partial agonists for stimulating cAMP accumulation (50 and 57% of the response to isoproterenol, respectively). The potency of polibegron was 1.71- and 4.5-fold higher than that of isoproterenol and BRL37344, respectively. These results indicate that polibegron acts as a potent, but partial, agonist at human β3ARs. In C57BL/6N mice with obesity induced by a high-fat diet, similar effects of the equimolar intraperitoneal administration of polibegron and BRL37344 were observed on weight, visceral fat and plasma levels of glucose, cholesterol and triglycerides. Similarities and differences between species related to ligand-receptor interactions can be useful for drug designing.
Collapse
Affiliation(s)
- Marvin A Soriano-Ursúa
- Departamentos de Fisiología, Bioquímica y Sección de Estudios de Posgrado E Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340, Mexico City, Mexico.
| | - José-Antonio Arias-Montaño
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del I.P.N., Av. IPN 2508, 07360, Mexico City, Mexico
| | - Ana-Lilia Ocampo-Néstor
- Departamentos de Fisiología, Bioquímica y Sección de Estudios de Posgrado E Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340, Mexico City, Mexico
- Departamento de Nefrología, Hospital General de México "Dr. Eduardo Liceaga", Dr. Balmis 148, Alc. Cuauhtémoc, 06720, Mexico City, Mexico
| | - Christian F Hernández-Martínez
- Departamentos de Fisiología, Bioquímica y Sección de Estudios de Posgrado E Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340, Mexico City, Mexico
| | - Iván Santillán-Torres
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del I.P.N., Av. IPN 2508, 07360, Mexico City, Mexico
| | - Erik Andrade-Jorge
- Departamentos de Fisiología, Bioquímica y Sección de Estudios de Posgrado E Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340, Mexico City, Mexico
| | - Rafael Valdez-Ortiz
- Departamento de Nefrología, Hospital General de México "Dr. Eduardo Liceaga", Dr. Balmis 148, Alc. Cuauhtémoc, 06720, Mexico City, Mexico
| | - Cecilia Fernández-Del Valle
- Área de Investigación Médica, Productos Medix, S.A. de C.V., Calzada del Hueso 39, Ejido Viejo Santa Úrsula Coapa, Coyoacán, 04650, Mexico City, Mexico
| | - José G Trujillo-Ferrara
- Departamentos de Fisiología, Bioquímica y Sección de Estudios de Posgrado E Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340, Mexico City, Mexico.
| |
Collapse
|
12
|
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: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
13
|
Sarsani V, Brotman SM, Xianyong Y, Fernandes Silva L, Laakso M, Spracklen CN. A cross-ancestry genome-wide meta-analysis, fine-mapping, and gene prioritization approach to characterize the genetic architecture of adiponectin. HGG ADVANCES 2024; 5:100252. [PMID: 37859345 PMCID: PMC10652123 DOI: 10.1016/j.xhgg.2023.100252] [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: 06/29/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023] Open
Abstract
Previous genome-wide association studies (GWASs) for adiponectin, a complex trait linked to type 2 diabetes and obesity, identified >20 associated loci. However, most loci were identified in populations of European ancestry, and many of the target genes underlying the associations remain unknown. We conducted a cross-ancestry adiponectin GWAS meta-analysis in ≤46,434 individuals from the Metabolic Syndrome in Men (METSIM) cohort and the ADIPOGen and AGEN consortiums. We combined study-specific association summary statistics using a fixed-effects, inverse variance-weighted approach. We identified 22 loci associated with adiponectin (p < 5×10-8), including 15 known and seven previously unreported loci. Among individuals of European ancestry, Genome-wide Complex Traits Analysis joint conditional analysis (GCTA-COJO) identified 14 additional distinct signals at the ADIPOQ, CDH13, HCAR1, and ZNF664 loci. Leveraging the cross-ancestry data, FINEMAP + SuSiE identified 45 causal variants (PP > 0.9), which also exhibited potential pleiotropy for cardiometabolic traits. To prioritize target genes at associated loci, we propose a combinatorial likelihood scoring formalism (Gene Priority Score [GPScore]) based on measures derived from 11 gene prioritization strategies and the physical distance to the transcription start site. With GPScore, we prioritize the 30 most probable target genes underlying the adiponectin-associated variants in the cross-ancestry analysis, including well-known causal genes (e.g., ADIPOQ, CDH13) and additional genes (e.g., CSF1, RGS17). Functional association networks revealed complex interactions of prioritized genes, their functionally connected genes, and their underlying pathways centered around insulin and adiponectin signaling, indicating an essential role in regulating energy balance in the body, inflammation, coagulation, fibrinolysis, insulin resistance, and diabetes. Overall, our analyses identify and characterize adiponectin association signals and inform experimental interrogation of target genes for adiponectin.
Collapse
Affiliation(s)
- Vishal Sarsani
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA, USA
| | - Sarah M Brotman
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yin Xianyong
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Lillian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Cassandra N Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA.
| |
Collapse
|
14
|
Ahles A, Engelhardt S. Genetic Variants of Adrenoceptors. Handb Exp Pharmacol 2024; 285:27-54. [PMID: 37578621 DOI: 10.1007/164_2023_676] [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] [Indexed: 08/15/2023]
Abstract
Adrenoceptors are class A G-protein-coupled receptors grouped into three families (α1-, α2-, and β-adrenoceptors), each one including three members. All nine corresponding adrenoceptor genes display genetic variation in their coding and adjacent non-coding genomic region. Coding variants, i.e., nucleotide exchanges within the transcribed and translated receptor sequence, may result in a difference in amino acid sequence thus altering receptor function and signaling. Such variants have been intensely studied in vitro in overexpression systems and addressed in candidate-gene studies for distinct clinical parameters. In recent years, large cohorts were analyzed in genome-wide association studies (GWAS), where variants are detected as significant in context with specific traits. These studies identified two of the in-depth characterized 18 coding variants in adrenoceptors as repeatedly statistically significant genetic risk factors - p.Arg389Gly in the β1- and p.Thr164Ile in the β2-adrenoceptor, along with 56 variants in the non-coding regions adjacent to the adrenoceptor gene loci, the functional role of which is largely unknown at present. This chapter summarizes current knowledge on the two coding variants in adrenoceptors that have been consistently validated in GWAS and provides a prospective overview on the numerous non-coding variants more recently attributed to adrenoceptor gene loci.
Collapse
Affiliation(s)
- Andrea Ahles
- Institute of Pharmacology and Toxicology, Technical University of Munich (TUM), Munich, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University of Munich (TUM), Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
15
|
Dwaib HS, Michel MC. Is the β 3-Adrenoceptor a Valid Target for the Treatment of Obesity and/or Type 2 Diabetes? Biomolecules 2023; 13:1714. [PMID: 38136585 PMCID: PMC10742325 DOI: 10.3390/biom13121714] [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: 10/09/2023] [Revised: 11/21/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
β3-Adrenoceptors mediate several functions in rodents that could be beneficial for the treatment of obesity and type 2 diabetes. This includes promotion of insulin release from the pancreas, cellular glucose uptake, lipolysis, and thermogenesis in brown adipose tissue. In combination, they lead to a reduction of body weight in several rodent models including ob/ob mice and Zucker diabetic fatty rats. These findings stimulated drug development programs in various pharmaceutical companies, and at least nine β3-adrenoceptor agonists have been tested in clinical trials. However, all of these projects were discontinued due to the lack of clinically relevant changes in body weight. Following a concise historical account of discoveries leading to such drug development programs we discuss species differences that explain why β3-adrenoceptors are not a meaningful drug target for the treatment of obesity and type 2 diabetes in humans.
Collapse
Affiliation(s)
- Haneen S. Dwaib
- Department of Clinical Nutrition and Dietetics, Palestine Ahliya University, Bethlehem P.O. Box 1041, Palestine;
| | - Martin C. Michel
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
16
|
Robinson EL, Bagchi RA, Major JL, Bergman BC, Matsuda JL, McKinsey TA. HDAC11 inhibition triggers bimodal thermogenic pathways to circumvent adipocyte catecholamine resistance. J Clin Invest 2023; 133:e168192. [PMID: 37607030 PMCID: PMC10541202 DOI: 10.1172/jci168192] [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: 12/19/2022] [Accepted: 08/03/2023] [Indexed: 08/24/2023] Open
Abstract
Stimulation of adipocyte β-adrenergic receptors (β-ARs) induces expression of uncoupling protein 1 (UCP1), promoting nonshivering thermogenesis. Association of β-ARs with a lysine-myristoylated form of A kinase-anchoring protein 12 (AKAP12, also known as gravin-α) is required for downstream signaling that culminates in UCP1 induction. Conversely, demyristoylation of gravin-α by histone deacetylase 11 (HDAC11) suppresses this pathway. Whether inhibition of HDAC11 in adipocytes is sufficient to drive UCP1 expression independently of β-ARs is not known. Here, we demonstrate that adipocyte-specific deletion of HDAC11 in mice leads to robust induction of UCP1 in adipose tissue (AT), resulting in increased body temperature. These effects are mimicked by treating mice in vivo or human AT ex vivo with an HDAC11-selective inhibitor, FT895. FT895 triggers biphasic, gravin-α myristoylation-dependent induction of UCP1 protein expression, with a noncanonical acute response that is posttranscriptional and independent of protein kinase A (PKA), and a delayed response requiring PKA activity and new Ucp1 mRNA synthesis. Remarkably, HDAC11 inhibition promotes UCP1 expression even in models of adipocyte catecholamine resistance where β-AR signaling is blocked. These findings define cell-autonomous, multimodal roles for HDAC11 as a suppressor of thermogenesis, and highlight the potential of inhibiting HDAC11 to therapeutically alter AT phenotype independently of β-AR stimulation.
Collapse
Affiliation(s)
- Emma L. Robinson
- Department of Medicine, Division of Cardiology
- Consortium for Fibrosis Research & Translation, and
| | - Rushita A. Bagchi
- Department of Medicine, Division of Cardiology
- Consortium for Fibrosis Research & Translation, and
| | - Jennifer L. Major
- Department of Medicine, Division of Cardiology
- Consortium for Fibrosis Research & Translation, and
| | - Bryan C. Bergman
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jennifer L. Matsuda
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA
| | - Timothy A. McKinsey
- Department of Medicine, Division of Cardiology
- Consortium for Fibrosis Research & Translation, and
| |
Collapse
|
17
|
Lan Q, Liufu S, Liu X, Ai N, Xu X, Li X, Yu Z, Yin Y, Liu M, Ma H. Comprehensive analysis of transcriptomic and metabolomic profiles uncovered the age-induced dynamic development pattern of subcutaneous fat in Ningxiang pig. Gene 2023; 880:147624. [PMID: 37422178 DOI: 10.1016/j.gene.2023.147624] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Enhancing meat production and quality is the eternal theme for pig breeding industries. Fat deposition has always been the focus of research in practical production because it is closely linked to pig production efficiency and pork quality. In the current study, multi-omics techniques were performed to explore the modulatory mechanisms of backfat (BF) accumulation at three core developmental stages for Ningxiang pigs. Our results identified that 15 differentially expressed genes (DEGs) and 9 significantly changed metabolites (SCMs) contributed to the BF development via the cAMP signaling pathway, regulation of lipolysis in adipocytes, and biosynthesis of unsaturated fatty acids. Herein, we found a series of candidate genes such as adrenoceptor beta 1 (ADRB1), adenylate cyclase 5 (ADCY5), ATPase Na+/K+ transporting subunit beta 1 (ATP1B1), ATPase plasma membrane Ca2+ transporting 3 (ATP2B3), ATPase Na+/K+ transporting subunit alpha 2 (ATP1A2), perilipin 1 (PLIN1), patatin like phospholipase domain containing 3 (PNPLA3), ELOVL fatty acid elongase 5 (ELOVL5) and metabolites like epinephrine, cAMP, arachidonic acid, oleic acid, linoleic acid, and docosahexaenoic acid existed age-specificeffects and played important roles in lipolysis, fat accumulation, and fatty acid composition. Our findings provide a reference for molecular mechanisms in BF tissue development and the optimization of carcass quality.
Collapse
Affiliation(s)
- Qun Lan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, PR China
| | - Sui Liufu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, PR China
| | - Xiaolin Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, PR China
| | - Nini Ai
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, PR China
| | - Xueli Xu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, PR China
| | - Xintong Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, PR China
| | - Zonggang Yu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, PR China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, PR China
| | - Mei Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, PR China.
| | - Haiming Ma
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, PR China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, PR China.
| |
Collapse
|
18
|
Jagtap U, Paul A. UCP1 activation: Hottest target in the thermogenesis pathway to treat obesity using molecules of synthetic and natural origin. Drug Discov Today 2023; 28:103717. [PMID: 37467882 DOI: 10.1016/j.drudis.2023.103717] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Uncoupling protein 1 (UCP1) has been discovered as a possible target for obesity treatment because of its widespread distribution in the inner mitochondrial membrane of brown adipose tissue (BAT) and high energy expenditure capabilities to burn calories as heat. UCP1 is dormant and does not produce heat without activation as it is inhibited by purine nucleotides. However, activation of UCP1 via either direct interaction with the UCP1 protein, an increase in the expression of UCP1 genes or the physiological production of fatty acids can lead to a rise in the thermogenesis phenomenon. Hence, activation of UCP1 through small molecules of synthetic and natural origin can be considered as a promising strategy to mitigate obesity.
Collapse
Affiliation(s)
- Utkarsh Jagtap
- Laboratory of Natural Product Chemistry, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Atish Paul
- Laboratory of Natural Product Chemistry, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
19
|
Wang S, Liu Y, Chen J, He Y, Ma W, Liu X, Sun X. Effects of multi-organ crosstalk on the physiology and pathology of adipose tissue. Front Endocrinol (Lausanne) 2023; 14:1198984. [PMID: 37383400 PMCID: PMC10293893 DOI: 10.3389/fendo.2023.1198984] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/26/2023] [Indexed: 06/30/2023] Open
Abstract
In previous studies, adipocytes were found to play an important role in regulating whole-body nutrition and energy balance, and are also important in energy metabolism, hormone secretion, and immune regulation. Different adipocytes have different contributions to the body, with white adipocytes primarily storing energy and brown adipocytes producing heat. Recently discovered beige adipocytes, which have characteristics in between white and brown adipocytes, also have the potential to produce heat. Adipocytes interact with other cells in the microenvironment to promote blood vessel growth and immune and neural network interactions. Adipose tissue plays an important role in obesity, metabolic syndrome, and type 2 diabetes. Dysfunction in adipose tissue endocrine and immune regulation can cause and promote the occurrence and development of related diseases. Adipose tissue can also secrete multiple cytokines, which can interact with organs; however, previous studies have not comprehensively summarized the interaction between adipose tissue and other organs. This article reviews the effect of multi-organ crosstalk on the physiology and pathology of adipose tissue, including interactions between the central nervous system, heart, liver, skeletal muscle, and intestines, as well as the mechanisms of adipose tissue in the development of various diseases and its role in disease treatment. It emphasizes the importance of a deeper understanding of these mechanisms for the prevention and treatment of related diseases. Determining these mechanisms has enormous potential for identifying new targets for treating diabetes, metabolic disorders, and cardiovascular diseases.
Collapse
Affiliation(s)
- Sufen Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yifan Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jiaqi Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yuejing He
- Clinical Laboratory, Dongguan Eighth People’s Hospital, Dongguan, China
| | - Wanrui Ma
- Department of General Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Xinguang Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Xuerong Sun
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan, China
| |
Collapse
|
20
|
Michel LYM. Extracellular Vesicles in Adipose Tissue Communication with the Healthy and Pathological Heart. Int J Mol Sci 2023; 24:ijms24097745. [PMID: 37175451 PMCID: PMC10177965 DOI: 10.3390/ijms24097745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/11/2023] [Accepted: 04/16/2023] [Indexed: 05/15/2023] Open
Abstract
Adipose tissue and its diverse cell types constitute one of the largest endocrine organs. With multiple depot locations, adipose tissue plays an important regulatory role through paracrine and endocrine communication, particularly through the secretion of a wide range of bioactive molecules, such as nucleic acids, proteins, lipids or adipocytokines. Over the past several years, research has uncovered a myriad of interorgan communication signals mediated by small lipid-derived nanovesicles known as extracellular vesicles (EVs), in which secreted bioactive molecules are stably transported as cargo molecules and delivered to adjacent cells or remote organs. EVs constitute an essential part of the human adipose secretome, and there is a growing body of evidence showing the crucial implications of adipose-derived EVs in the regulation of heart function and its adaptative capacity. The adipose tissue modifications and dysfunction observed in obesity and aging tremendously affect the adipose-EV secretome, with important consequences for the myocardium. The present review presents a comprehensive analysis of the findings in this novel area of research, reports the key roles played by adipose-derived EVs in interorgan cross-talk with the heart and discusses their implications in physiological and pathological conditions affecting adipose tissue and/or the heart (pressure overload, ischemia, diabetic cardiomyopathy, etc.).
Collapse
Affiliation(s)
- Lauriane Y M Michel
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 57 Avenue Hippocrate, 1200 Brussels, Belgium
| |
Collapse
|
21
|
Coulter AA, Greenway FL, Zhang D, Ghosh S, Coulter CR, James SL, He Y, Cusimano LA, Rebello CJ. Naringenin and β-carotene convert human white adipocytes to a beige phenotype and elevate hormone- stimulated lipolysis. Front Endocrinol (Lausanne) 2023; 14:1148954. [PMID: 37143734 PMCID: PMC10153092 DOI: 10.3389/fendo.2023.1148954] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/20/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction Naringenin, a peroxisome proliferator-activated receptor (PPAR) activator found in citrus fruits, upregulates markers of thermogenesis and insulin sensitivity in human adipose tissue. Our pharmacokinetics clinical trial demonstrated that naringenin is safe and bioavailable, and our case report showed that naringenin causes weight loss and improves insulin sensitivity. PPARs form heterodimers with retinoic-X-receptors (RXRs) at promoter elements of target genes. Retinoic acid is an RXR ligand metabolized from dietary carotenoids. The carotenoid β-carotene reduces adiposity and insulin resistance in clinical trials. Our goal was to examine if carotenoids strengthen the beneficial effects of naringenin on human adipocyte metabolism. Methods Human preadipocytes from donors with obesity were differentiated in culture and treated with 8µM naringenin + 2µM β-carotene (NRBC) for seven days. Candidate genes involved in thermogenesis and glucose metabolism were measured as well as hormone-stimulated lipolysis. Results We found that β-carotene acts synergistically with naringenin to boost UCP1 and glucose metabolism genes including GLUT4 and adiponectin, compared to naringenin alone. Protein levels of PPARα, PPARγ and PPARγ-coactivator-1α, key modulators of thermogenesis and insulin sensitivity, were also upregulated after treatment with NRBC. Transcriptome sequencing was conducted and the bioinformatics analyses of the data revealed that NRBC induced enzymes for several non-UCP1 pathways for energy expenditure including triglyceride cycling, creatine kinases, and Peptidase M20 Domain Containing 1 (PM20D1). A comprehensive analysis of changes in receptor expression showed that NRBC upregulated eight receptors that have been linked to lipolysis or thermogenesis including the β1-adrenergic receptor and the parathyroid hormone receptor. NRBC increased levels of triglyceride lipases and agonist-stimulated lipolysis in adipocytes. We observed that expression of RXRγ, an isoform of unknown function, was induced ten-fold after treatment with NRBC. We show that RXRγ is a coactivator bound to the immunoprecipitated PPARγ protein complex from white and beige human adipocytes. Discussion There is a need for obesity treatments that can be administered long-term without side effects. NRBC increases the abundance and lipolytic response of multiple receptors for hormones released after exercise and cold exposure. Lipolysis provides the fuel for thermogenesis, and these observations suggest that NRBC has therapeutic potential.
Collapse
Affiliation(s)
- Ann A. Coulter
- Computational Biology, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Frank L. Greenway
- Clinical Trials, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Dachuan Zhang
- Biostatistics, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Sujoy Ghosh
- Adjunct Faculty, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Cathryn R. Coulter
- Computational Biology, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Sarah L. James
- Computational Biology, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Yanlin He
- Brain Glycemic and Metabolism Control, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Luke A. Cusimano
- Cusimano Plastic and Reconstructive Surgery, Baton Rouge, LA, United States
| | - Candida J. Rebello
- Nutrition and Chronic Disease, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| |
Collapse
|
22
|
Robinson EL, Bagchi RA, Major JL, Bergman BC, Madsuda JL, McKinsey TA. HDAC11 inhibition triggers bimodal thermogenic pathways to circumvent adipocyte catecholamine resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.29.534830. [PMID: 37034582 PMCID: PMC10081236 DOI: 10.1101/2023.03.29.534830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Stimulation of adipocyte β-adrenergic receptors (β-ARs) induces expression of uncoupling protein 1 (UCP1), promoting non-shivering thermogenesis. Association of β-ARs with a lysine myristoylated form of A-kinase anchoring protein 12 (AKAP12)/gravin-α is required for downstream signaling that culminates in UCP1 induction. Conversely, demyristoylation of gravin-α by histone deacetylase 11 (HDAC11) suppresses this pathway. Whether inhibition of HDAC11 in adipocytes is sufficient to drive UCP1 expression independently of β-ARs is not known. Here, we demonstrate that adipocyte-specific deletion of HDAC11 in mice leads to robust induction of UCP1 in adipose tissue (AT), resulting in increased body temperature. These effects are mimicked by treating mice in vivo or human AT ex vivo with an HDAC11-selective inhibitor, FT895. FT895 triggers biphasic, gravin-α myristoylation-dependent induction of UCP1 protein expression, with a non-canonical acute response that is post-transcriptional and independent of protein kinase A (PKA), and a delayed response requiring PKA activity and new Ucp1 mRNA synthesis. Remarkably, HDAC11 inhibition promotes UCP1 expression even in models of adipocyte catecholamine resistance where β-AR signaling is blocked. These findings define cell autonomous, multi-modal roles for HDAC11 as a suppressor of thermogenesis, and highlight the potential of inhibiting HDAC11 to therapeutically alter AT phenotype independently of β-AR stimulation.
Collapse
Affiliation(s)
- Emma L. Robinson
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-2507; USA
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-2507; USA
| | - Rushita A. Bagchi
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-2507; USA
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-2507; USA
| | - Jennifer L. Major
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-2507; USA
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-2507; USA
| | - Bryan C. Bergman
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-2507; USA
| | - Jennifer L. Madsuda
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA
| | - Timothy A. McKinsey
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-2507; USA
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-2507; USA
| |
Collapse
|
23
|
Abstract
Brown adipose tissue (BAT) displays the unique capacity to generate heat through uncoupled oxidative phosphorylation that makes it a very attractive therapeutic target for cardiometabolic diseases. Here, we review BAT cellular metabolism, its regulation by the central nervous and endocrine systems and circulating metabolites, the plausible roles of this tissue in human thermoregulation, energy balance, and cardiometabolic disorders, and the current knowledge on its pharmacological stimulation in humans. The current definition and measurement of BAT in human studies relies almost exclusively on BAT glucose uptake from positron emission tomography with 18F-fluorodeoxiglucose, which can be dissociated from BAT thermogenic activity, as for example in insulin-resistant states. The most important energy substrate for BAT thermogenesis is its intracellular fatty acid content mobilized from sympathetic stimulation of intracellular triglyceride lipolysis. This lipolytic BAT response is intertwined with that of white adipose (WAT) and other metabolic tissues, and cannot be independently stimulated with the drugs tested thus far. BAT is an interesting and biologically plausible target that has yet to be fully and selectively activated to increase the body's thermogenic response and shift energy balance. The field of human BAT research is in need of methods able to directly, specifically, and reliably measure BAT thermogenic capacity while also tracking the related thermogenic responses in WAT and other tissues. Until this is achieved, uncertainty will remain about the role played by this fascinating tissue in human cardiometabolic diseases.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | | | - Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec City, Quebec, G1V 4G5, Canada
| |
Collapse
|
24
|
Straat ME, Hoekx CA, van Velden FHP, Pereira Arias-Bouda LM, Dumont L, Blondin DP, Boon MR, Martinez-Tellez B, Rensen PCN. Stimulation of the beta-2-adrenergic receptor with salbutamol activates human brown adipose tissue. Cell Rep Med 2023; 4:100942. [PMID: 36812890 PMCID: PMC9975328 DOI: 10.1016/j.xcrm.2023.100942] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/30/2022] [Accepted: 01/20/2023] [Indexed: 02/23/2023]
Abstract
While brown adipose tissue (BAT) is activated by the beta-3-adrenergic receptor (ADRB3) in rodents, in human brown adipocytes, the ADRB2 is dominantly present and responsible for noradrenergic activation. Therefore, we performed a randomized double-blinded crossover trial in young lean men to compare the effects of single intravenous bolus of the ADRB2 agonist salbutamol without and with the ADRB1/2 antagonist propranolol on glucose uptake by BAT, assessed by dynamic 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography-computed tomography scan (i.e., primary outcome). Salbutamol, compared with salbutamol with propranolol, increases glucose uptake by BAT, without affecting the glucose uptake by skeletal muscle and white adipose tissue. The salbutamol-induced glucose uptake by BAT positively associates with the increase in energy expenditure. Notably, participants with high salbutamol-induced glucose uptake by BAT have lower body fat mass, waist-hip ratio, and serum LDL-cholesterol concentration. In conclusion, specific ADRB2 agonism activates human BAT, which warrants investigation of ADRB2 activation in long-term studies (EudraCT: 2020-004059-34).
Collapse
Affiliation(s)
- Maaike E Straat
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Carlijn A Hoekx
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Floris H P van Velden
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Lenka M Pereira Arias-Bouda
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Lauralyne Dumont
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; Department of Physiology-Pharmacology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Denis P Blondin
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; Department of Medicine, Division of Neurology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Mariëtte R Boon
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Borja Martinez-Tellez
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
25
|
GPCR in Adipose Tissue Function-Focus on Lipolysis. Biomedicines 2023; 11:biomedicines11020588. [PMID: 36831123 PMCID: PMC9953751 DOI: 10.3390/biomedicines11020588] [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: 01/20/2023] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Adipose tissue can be divided anatomically, histologically, and functionally into two major entities white and brown adipose tissues (WAT and BAT, respectively). WAT is the primary energy depot, storing most of the bioavailable triacylglycerol molecules of the body, whereas BAT is designed for dissipating energy in the form of heat, a process also known as non-shivering thermogenesis as a defense against a cold environment. Importantly, BAT-dependent energy dissipation directly correlates with cardiometabolic health and has been postulated as an intriguing target for anti-obesity therapies. In general, adipose tissue (AT) lipid content is defined by lipid uptake and lipogenesis on one side, and, on the other side, it is defined by the breakdown of lipids and the release of fatty acids by lipolysis. The equilibrium between lipogenesis and lipolysis is important for adipocyte and general metabolic homeostasis. Overloading adipocytes with lipids causes cell stress, leading to the recruitment of immune cells and adipose tissue inflammation, which can affect the whole organism (metaflammation). The most important consequence of energy and lipid overload is obesity and associated pathophysiologies, including insulin resistance, type 2 diabetes, and cardiovascular disease. The fate of lipolysis products (fatty acids and glycerol) largely differs between AT: WAT releases fatty acids into the blood to deliver energy to other tissues (e.g., muscle). Activation of BAT, instead, liberates fatty acids that are used within brown adipocyte mitochondria for thermogenesis. The enzymes involved in lipolysis are tightly regulated by the second messenger cyclic adenosine monophosphate (cAMP), which is activated or inhibited by G protein-coupled receptors (GPCRs) that interact with heterotrimeric G proteins (G proteins). Thus, GPCRs are the upstream regulators of the equilibrium between lipogenesis and lipolysis. Moreover, GPCRs are of special pharmacological interest because about one third of the approved drugs target GPCRs. Here, we will discuss the effects of some of most studied as well as "novel" GPCRs and their ligands. We will review different facets of in vitro, ex vivo, and in vivo studies, obtained with both pharmacological and genetic approaches. Finally, we will report some possible therapeutic strategies to treat obesity employing GPCRs as primary target.
Collapse
|
26
|
Wang CJ, Noble PB, Elliot JG, James AL, Wang KCW. From Beneath the Skin to the Airway Wall: Understanding the Pathological Role of Adipose Tissue in Comorbid Asthma-Obesity. Compr Physiol 2023; 13:4321-4353. [PMID: 36715283 DOI: 10.1002/cphy.c220011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This article provides a contemporary report on the role of adipose tissue in respiratory dysfunction. Adipose tissue is distributed throughout the body, accumulating beneath the skin (subcutaneous), around organs (visceral), and importantly in the context of respiratory disease, has recently been shown to accumulate within the airway wall: "airway-associated adipose tissue." Excessive adipose tissue deposition compromises respiratory function and increases the severity of diseases such as asthma. The mechanisms of respiratory impairment are inflammatory, structural, and mechanical in nature, vary depending on the anatomical site of deposition and adipose tissue subtype, and likely contribute to different phenotypes of comorbid asthma-obesity. An understanding of adipose tissue-driven pathophysiology provides an opportunity for diagnostic advancement and patient-specific treatment. As an exemplar, the potential impact of airway-associated adipose tissue is highlighted, and how this may change the management of a patient with asthma who is also obese. © 2023 American Physiological Society. Compr Physiol 13:4321-4353, 2023.
Collapse
Affiliation(s)
- Carolyn J Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - John G Elliot
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Kimberley C W Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
27
|
Thermogenic Adipose Redox Mechanisms: Potential Targets for Metabolic Disease Therapies. Antioxidants (Basel) 2023; 12:antiox12010196. [PMID: 36671058 PMCID: PMC9854447 DOI: 10.3390/antiox12010196] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/07/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Metabolic diseases, such as diabetes and non-alcoholic fatty liver disease (NAFLD), have several negative health outcomes on affected humans. Dysregulated energy metabolism is a key component underlying the pathophysiology of these conditions. Adipose tissue is a fundamental regulator of energy homeostasis that utilizes several redox reactions to carry out the metabolism. Brown and beige adipose tissues, in particular, perform highly oxidative reactions during non-shivering thermogenesis to dissipate energy as heat. The appropriate regulation of energy metabolism then requires coordinated antioxidant mechanisms to counterbalance the oxidation reactions. Indeed, non-shivering thermogenesis activation can cause striking changes in concentrations of both oxidants and antioxidants in order to adapt to various oxidative environments. Current therapeutic options for metabolic diseases either translate poorly from rodent models to humans (in part due to the challenges of creating a physiologically relevant rodent model) or tend to have numerous side effects, necessitating novel therapies. As increased brown adipose tissue activity results in enhanced energy expenditure and is associated with beneficial effects on metabolic health, such as decreased obesity, it has gathered great interest as a modulator of metabolic disease. One potential reason for the beneficial health effects may be that although non-shivering thermogenesis is enormously oxidative, it is also associated with decreased oxidant formation after its activation. However, targeting its redox mechanisms specifically to alter metabolic disease remains an underexplored area. Therefore, this review will discuss the role of adipose tissue in energy homeostasis, non-shivering thermogenesis in adults, and redox mechanisms that may serve as novel therapeutic targets of metabolic disease.
Collapse
|
28
|
Chu DT, Bui NL, Le NH. Adrenoceptors and SCD1 in adipocytes/adipose tissues: The expression and variation in health and obesity. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 194:311-332. [PMID: 36631196 DOI: 10.1016/bs.pmbts.2022.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Obesity, considered a metabolic disorder, is one of the most significant health issues that the community has to cope with today. A rising number of studies have been conducted to find out promising genetic targets for obese treatment. The sympathetic nervous system was proven to possess remarkable roles in energy metabolism, including the stimulation of lipolysis as well as thermogenesis, via distinct adrenoceptors appearing on the membrane of adipocyte. A decrease of β-adrenoceptor expression has been observed in obese individuals, which is related to reducing energy expenditure and developing obesity. While that the deficiency of stearoyl-CoA desaturase-1 (SCD1), which is a promising target for treatments of metabolic diseases, decreases oxidation and promotes the synthesis of fatty acids. Here, we emphasized several differences between distinct adrenoceptor subtypes, including their mRNA expression level and function in white adipose tissue and brown adipose tissue. We also highlighted SCD1's roles related to the progression of adipocytes and its changing expression under the obese condition in both rodents and humans, and furthermore, tried to figure out the interaction between adrenoceptors and SCD1 in adipose tissue.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam.
| | - Nhat-Le Bui
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Ngoc Hoan Le
- Faculty of Biology, Hanoi National University of Education, Hanoi, Vietnam
| |
Collapse
|
29
|
Activation of β-Adrenoceptors Promotes Lipid Droplet Accumulation in MCF-7 Breast Cancer Cells via cAMP/PKA/EPAC Pathways. Int J Mol Sci 2023; 24:ijms24010767. [PMID: 36614209 PMCID: PMC9820888 DOI: 10.3390/ijms24010767] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Physiologically, β-adrenoceptors are major regulators of lipid metabolism, which may be reflected in alterations in lipid droplet dynamics. β-adrenoceptors have also been shown to participate in breast cancer carcinogenesis. Since lipid droplets may be seen as a hallmark of cancer, the present study aimed to investigate the role of β-adrenoceptors in the regulation of lipid droplet dynamics in MCF-7 breast cancer cells. Cells were treated for up to 72 h with adrenaline (an endogenous adrenoceptor agonist), isoprenaline (a non-selective β-adrenoceptor agonist) and salbutamol (a selective β2-selective agonist), and their effects on lipid droplets were evaluated using Nile Red staining. Adrenaline or isoprenaline, but not salbutamol, caused a lipid-accumulating phenotype in the MCF-7 cells. These effects were significantly reduced by selective β1- and β3-antagonists (10 nM atenolol and 100 nM L-748,337, respectively), indicating a dependence on both β1- and β3-adrenoceptors. These effects were dependent on the cAMP signalling pathway, involving both protein kinase A (PKA) and cAMP-dependent guanine-nucleotide-exchange (EPAC) proteins: treatment with cAMP-elevating agents (forskolin or 8-Br-cAMP) induced lipid droplet accumulation, whereas either 1 µM H-89 or 1 µM ESI-09 (PKA or EPAC inhibitors, respectively) abrogated this effect. Taken together, the present results demonstrate the existence of a β-adrenoceptor-mediated regulation of lipid droplet dynamics in breast cancer cells, likely involving β1- and β3-adrenoceptors, revealing a new mechanism by which adrenergic stimulation may influence cancer cell metabolism.
Collapse
|
30
|
Chen Y, Hu Q, Wang C, Wang T. The crosstalk between BAT thermogenesis and skeletal muscle dysfunction. Front Physiol 2023; 14:1132830. [PMID: 37153220 PMCID: PMC10160478 DOI: 10.3389/fphys.2023.1132830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
Metabolic defects increase the risk of skeletal muscle diseases, and muscle impairment might worsen metabolic disruption, leading to a vicious cycle. Both brown adipose tissue (BAT) and skeletal muscle play important roles in non-shivering thermogenesis to regulate energy homeostasis. BAT regulates body temperature, systemic metabolism, and seretion of batokines that have positive or negative impacts on skeletal muscle. Conversely, muscle can secrete myokines that regulate BAT function. This review explained the crosstalk between BAT and skeletal muscle, and then discussed the batokines and highlighted their impact on skeletal muscle under physiological conditions. BAT is now considered a potential therapeutic target for obesity and diabetes treatment. Moreover, manipulation of BAT may be an attractive approach for the treatment of muscle weakness by correcting metabolic deficits. Therefore, exploring BAT as a potential treatment for sarcopenia could be a promising avenue for future research.
Collapse
Affiliation(s)
- Yao Chen
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Hu
- Health Management Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Changyi Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Changyi Wang, ; Tiantian Wang,
| | - Tiantian Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Changyi Wang, ; Tiantian Wang,
| |
Collapse
|
31
|
Brain-to-BAT - and Back?: Crosstalk between the Central Nervous System and Thermogenic Adipose Tissue in Development and Therapy of Obesity. Brain Sci 2022; 12:brainsci12121646. [PMID: 36552107 PMCID: PMC9775239 DOI: 10.3390/brainsci12121646] [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: 11/04/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
The body of mammals harbors two distinct types of adipose tissue: while cells within the white adipose tissue (WAT) store surplus energy as lipids, brown adipose tissue (BAT) is nowadays recognized as the main tissue for transforming chemical energy into heat. This process, referred to as 'non-shivering thermogenesis', is facilitated by the uncoupling of the electron transport across mitochondrial membranes from ATP production. BAT-dependent thermogenesis acts as a safeguarding mechanism under reduced ambient temperature but also plays a critical role in metabolic and energy homeostasis in health and disease. In this review, we summarize the evolutionary structure, function and regulation of the BAT organ under neuronal and hormonal control and discuss its mutual interaction with the central nervous system. We conclude by conceptualizing how better understanding the multifaceted communicative links between the brain and BAT opens avenues for novel therapeutic approaches to treat obesity and related metabolic disorders.
Collapse
|
32
|
Never-homozygous genetic variants in healthy populations are potential recessive disease candidates. NPJ Genom Med 2022; 7:54. [PMID: 36075934 PMCID: PMC9458638 DOI: 10.1038/s41525-022-00322-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
The rapid pace with which genetic variants are now being determined means there is a pressing need to understand how they affect biological systems. Variants from healthy individuals have previously been used to study blood groups or HLA diversity and to identify genes that can apparently be nonfunctional in healthy people. These studies and others have observed a lower than expected frequency of homozygous individuals for potentially deleterious alleles, which would suggest that several of these alleles can lead to recessive disorders. Here we exploited this principle to hunt for potential disease variants in genomes from healthy people. We identified at least 108 exclusively heterozygous variants with evidence for an impact on biological function. We discuss several examples of candidate variants/genes including CCDC8, PANK3, RHD and NLRP12. Overall, the results suggest there are many, comparatively frequent, potentially lethal or disease-causing variants lurking in healthy human populations.
Collapse
|
33
|
Pinckard KM, Stanford KI. The Heartwarming Effect of Brown Adipose Tissue. Mol Pharmacol 2022; 102:460-471. [PMID: 34933905 PMCID: PMC9341250 DOI: 10.1124/molpharm.121.000328] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/27/2021] [Indexed: 11/22/2022] Open
Abstract
Brown adipose tissue (BAT) is a metabolically active tissue that improves glucose metabolism and protects against the development of type 2 diabetes and obesity. However, the role of BAT to improve cardiovascular health has only recently been investigated. In this review, we discuss multiple mechanisms through which both the thermogenic and endocrine functions of BAT mediate cardiac health. β-adrenergic stimulation activates the thermogenic function of BAT, resulting in reduced circulating lipids and glucose, and enhanced clearance of hepatic cholesterol-enriched remnants leading to reduced atherosclerotic region size. Additionally, the thermogenic role of BAT has been implicated in activation of the protein kinase B-extracellular-signal-regulated kinase (ERK) 1/2 pathway after myocardial infarction (MI), contributing to reduced injury size. The endocrine function of BAT has also been implicated to improve both systemic metabolic health and cardiac health. Specifically, the batokines fibroblast growth factor 21 (FGF21) and 12,13-diHOME improve cardiovascular health via reduced hypertension, hypertrophy and MI injury size (FGF21) or by directly improving cardiac function via calcium cycling (12,13-diHOME). Finally, we discuss relevant pharmacological treatment methods currently aiming to activate BAT, typically through sympathetic activation. SIGNIFICANCE STATEMENT: This mini-review discusses the role of BAT to improve cardiac health via thermogenic and endocrine effects in both rodents and humans and highlights the need for therapeutic methods which activate or mimic BAT activity.
Collapse
Affiliation(s)
- Kelsey M Pinckard
- Department of Physiology and Cell Biology (K.M.P., K.I.S.), Center for Diabetes and Metabolism Research Center, Dorothy M. Davis Heart and Lung Research Institute (K.M.P., K.I.S.), and Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio (K.I.S.)
| | - Kristin I Stanford
- Department of Physiology and Cell Biology (K.M.P., K.I.S.), Center for Diabetes and Metabolism Research Center, Dorothy M. Davis Heart and Lung Research Institute (K.M.P., K.I.S.), and Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio (K.I.S.)
| |
Collapse
|
34
|
Osorio-Conles Ó, Olbeyra R, Moizé V, Ibarzabal A, Giró O, Viaplana J, Jiménez A, Vidal J, de Hollanda A. Positive Effects of a Mediterranean Diet Supplemented with Almonds on Female Adipose Tissue Biology in Severe Obesity. Nutrients 2022; 14:nu14132617. [PMID: 35807797 PMCID: PMC9267991 DOI: 10.3390/nu14132617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 02/06/2023] Open
Abstract
It has been suggested that weight-loss-independent Mediterranean diet benefits on cardiometabolic health and diabetes prevention may be mediated, at least in part, through the modulation of white adipose tissue (WAT) biology. This study aimed to evaluate the short-term effects of a dietary intervention based on the Mediterranean diet supplemented with almonds (MDSA) on the main features of obesity-associated WAT dysfunction. A total of 38 women with obesity were randomly assigned to a 3-month intervention with MDSA versus continuation of their usual dietary pattern. Subcutaneous (SAT) and visceral adipose tissue (VAT) biopsies were obtained before and after the dietary intervention, and at the end of the study period, respectively. MDSA favored the abundance of small adipocytes in WAT. In SAT, the expression of angiogenesis genes increased after MDSA intervention. In VAT, the expression of genes implicated in adipogenesis, angiogenesis, autophagy and fatty acid usage was upregulated. In addition, a higher immunofluorescence staining for PPARG, CD31+ cells and M2-like macrophages and increased ADRB1 and UCP2 protein contents were found compared to controls. Changes in WAT correlated with a significant reduction in circulating inflammatory markers and LDL-cholesterol levels. These results support a protective effect of a Mediterranean diet supplemented with almonds on obesity-related WAT dysfunction.
Collapse
Affiliation(s)
- Óscar Osorio-Conles
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (Ó.O.-C.); (V.M.); (J.V.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.O.); (O.G.); (A.J.)
| | - Romina Olbeyra
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.O.); (O.G.); (A.J.)
| | - Violeta Moizé
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (Ó.O.-C.); (V.M.); (J.V.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.O.); (O.G.); (A.J.)
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Ainitze Ibarzabal
- Gastrointestinal Surgery Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain;
| | - Oriol Giró
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.O.); (O.G.); (A.J.)
| | - Judith Viaplana
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (Ó.O.-C.); (V.M.); (J.V.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.O.); (O.G.); (A.J.)
| | - Amanda Jiménez
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.O.); (O.G.); (A.J.)
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Josep Vidal
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (Ó.O.-C.); (V.M.); (J.V.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.O.); (O.G.); (A.J.)
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Correspondence: (J.V.); (A.d.H.); Tel.: +34-93-227-20-12 (J.V.); +34-93-227-98-46 (A.d.H.); Fax: +34-93-227-55-89 (J.V. & A.d.H.)
| | - Ana de Hollanda
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.O.); (O.G.); (A.J.)
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Correspondence: (J.V.); (A.d.H.); Tel.: +34-93-227-20-12 (J.V.); +34-93-227-98-46 (A.d.H.); Fax: +34-93-227-55-89 (J.V. & A.d.H.)
| |
Collapse
|
35
|
Equisetin is an anti-obesity candidate through targeting 11 β-HSD1. Acta Pharm Sin B 2022; 12:2358-2373. [PMID: 35646525 PMCID: PMC9136616 DOI: 10.1016/j.apsb.2022.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity is increasingly prevalent globally, searching for therapeutic agents acting on adipose tissue is of great importance. Equisetin (EQST), a meroterpenoid isolated from a marine sponge-derived fungus, has been reported to display antibacterial and antiviral activities. Here, we revealed that EQST displayed anti-obesity effects acting on adipose tissue through inhibiting adipogenesis in vitro and attenuating HFD-induced obesity in mice, doing so without affecting food intake, blood pressure or heart rate. We demonstrated that EQST inhibited the enzyme activity of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), a therapeutic target of obesity in adipose tissue. Anti-obesity properties of EQST were all offset by applying excessive 11β-HSD1's substrates and 11β-HSD1 inhibition through knockdown in vitro or 11β-HSD1 knockout in vivo. In the 11β-HSD1 bypass model constructed by adding excess 11β-HSD1 products, EQST's anti-obesity effects disappeared. Furthermore, EQST directly bond to 11β-HSD1 protein and presented remarkable better intensity on 11β-HSD1 inhibition and better efficacy on anti-obesity than known 11β-HSD1 inhibitor. Therefore, EQST can be developed into anti-obesity candidate compound, and this study may provide more clues for developing higher effective 11β-HSD1 inhibitors.
Collapse
|
36
|
Scheel AK, Espelage L, Chadt A. Many Ways to Rome: Exercise, Cold Exposure and Diet-Do They All Affect BAT Activation and WAT Browning in the Same Manner? Int J Mol Sci 2022; 23:ijms23094759. [PMID: 35563150 PMCID: PMC9103087 DOI: 10.3390/ijms23094759] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 02/08/2023] Open
Abstract
The discovery of functional brown adipose tissue (BAT) in adult humans and the possibility to recruit beige cells with high thermogenic potential within white adipose tissue (WAT) depots opened the field for new strategies to combat obesity and its associated comorbidities. Exercise training as well as cold exposure and dietary components are associated with the enhanced accumulation of metabolically-active beige adipocytes and BAT activation. Both activated beige and brown adipocytes increase their metabolic rate by utilizing lipids to generate heat via non-shivering thermogenesis, which is dependent on uncoupling protein 1 (UCP1) in the inner mitochondrial membrane. Non-shivering thermogenesis elevates energy expenditure and promotes a negative energy balance, which may ameliorate metabolic complications of obesity and Type 2 Diabetes Mellitus (T2DM) such as insulin resistance (IR) in skeletal muscle and adipose tissue. Despite the recent advances in pharmacological approaches to reduce obesity and IR by inducing non-shivering thermogenesis in BAT and WAT, the administered pharmacological compounds are often associated with unwanted side effects. Therefore, lifestyle interventions such as exercise, cold exposure, and/or specified dietary regimens present promising anchor points for future disease prevention and treatment of obesity and T2DM. The exact mechanisms where exercise, cold exposure, dietary interventions, and pharmacological treatments converge or rather diverge in their specific impact on BAT activation or WAT browning are difficult to determine. In the past, many reviews have demonstrated the mechanistic principles of exercise- and/or cold-induced BAT activation and WAT browning. In this review, we aim to summarize not only the current state of knowledge on the various mechanistic principles of diverse external stimuli on BAT activation and WAT browning, but also present their translational potential in future clinical applications.
Collapse
Affiliation(s)
- Anna K. Scheel
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Medical Faculty, Düsseldorf, Auf’m Hennekamp 65, 40225 Duesseldorf, Germany; (A.K.S.); (L.E.)
- German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, 85764 München, Germany
| | - Lena Espelage
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Medical Faculty, Düsseldorf, Auf’m Hennekamp 65, 40225 Duesseldorf, Germany; (A.K.S.); (L.E.)
- German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, 85764 München, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Medical Faculty, Düsseldorf, Auf’m Hennekamp 65, 40225 Duesseldorf, Germany; (A.K.S.); (L.E.)
- German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, 85764 München, Germany
- Correspondence: ; Tel./Fax: +49-211-3382-577/430
| |
Collapse
|
37
|
Johansen OS, Ma T, Gerhart-Hines Z. Leveraging GPCR signaling in thermogenic fat to counteract metabolic diseases. Mol Metab 2022; 60:101474. [PMID: 35339729 PMCID: PMC9046952 DOI: 10.1016/j.molmet.2022.101474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/24/2022] [Accepted: 03/11/2022] [Indexed: 11/05/2022] Open
Abstract
Background Thermogenic brown and beige adipocytes are recognized for their unique capacity to consume extraordinary levels of metabolites and lipids from the blood to fuel heat-producing catabolic processes [[1], [2], [3], [4], [5], [6], [7]]. In humans, the functions of thermogenic adipocytes are associated with cardiometabolic protection and improved glycemic control [[8], [9], [10], [11], [12], [13]]. Consequently, engaging these macronutrient-consuming and energy-dissipating activities has gained attention as a promising therapeutic strategy for counteracting metabolic diseases, such as obesity and diabetes. Scope of review In this review, we highlight new advances in our understanding of the physiological role of G protein-coupled receptors (GPCRs) in controlling thermogenic adipocyte biology. We further extend our discussion to the opportunities and challenges posed by pharmacologically targeting different elements of GPCR signaling in these highly specialized fat cells. Major conclusions GPCRs represent appealing candidates through which to harness adipose thermogenesis. Yet safely and effectively targeting these druggable receptors on brown and beige adipocytes has thus far proven challenging. Therefore, continued interrogation across the GPCR landscape is necessary for future leaps within the field of thermogenic fat biology to unlock the therapeutic potential of adipocyte catabolism. Brown and beige thermogenic adipocytes robustly consume and catabolize macronutrients. The catabolic activity of thermogenic adipocytes promotes organismal energy balance. Thermogenic adipocyte functions are tightly controlled by G protein-coupled receptors (GPCRs). GPCRs can be potentially targeted at multiple levels to therapeutically harness thermogenic activity.
Collapse
Affiliation(s)
- Olivia Sveidahl Johansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, DK
| | - Tao Ma
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, DK; Embark Biotech ApS, Copenhagen, DK
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, DK; Embark Biotech ApS, Copenhagen, DK; Center for Adipocyte Signaling, Odense, DK.
| |
Collapse
|
38
|
Li Y, Fromme T. Uncoupling Protein 1 Does Not Produce Heat without Activation. Int J Mol Sci 2022; 23:2406. [PMID: 35269549 PMCID: PMC8910648 DOI: 10.3390/ijms23052406] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial uncoupling protein 1 (UCP1) is the crucial mechanistic component of heat production in classical brown fat and the newly identified beige or brite fat. Thermogenesis inevitably comes at a high energetic cost and brown fat, ultimately, is an energy-wasting organ. A constrained strategy that minimizes brown fat activity unless obligate will have been favored during natural selection to safeguard metabolic thriftiness. Accordingly, UCP1 is constitutively inhibited and is inherently not leaky without activation. It follows that increasing brown adipocyte number or UCP1 abundance genetically or pharmacologically does not lead to an automatic increase in thermogenesis or subsequent metabolic consequences in the absence of a plausible route of concomitant activation. Despite its apparent obviousness, this tenet is frequently ignored. Consequently, incorrect conclusions are often drawn from increased BAT or brite/beige depot mass, e.g., predicting or causally linking beneficial metabolic effects. Here, we highlight the inherently inactive nature of UCP1, with a particular emphasis on the molecular brakes and releases of UCP1 activation under physiological conditions. These controls of UCP1 activity represent potential targets of therapeutic interventions to unlock constraints and efficiently harness the energy-expending potential of brown fat to prevent and treat obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Yongguo Li
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Tobias Fromme
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| |
Collapse
|
39
|
Osorio-Conles Ó, Vega-Beyhart A, Ibarzabal A, Balibrea JM, Vidal J, de Hollanda A. Biological Determinants of Metabolic Syndrome in Visceral and Subcutaneous Adipose Tissue from Severely Obese Women. Int J Mol Sci 2022; 23:ijms23042394. [PMID: 35216509 PMCID: PMC8878297 DOI: 10.3390/ijms23042394] [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: 01/05/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 11/16/2022] Open
Abstract
The metabolic syndrome (MetS) is a cluster of the most dangerous heart attack risk factors: diabetes or raised fasting plasma glucose, abdominal obesity, high cholesterol and high blood pressure. The goal of this study is to compare the state of the main features of obesity-associated white adipose tissue (WAT) dysfunction in 66 women with severe obesity without (MetS-) or with MetS (MetS+). Fat cell area, adipocyte size distribution and histological fibrosis were analysed in visceral (VAT) and abdominal subcutaneous WAT (SAT) in 33 age- and BMI-matched pairs of MetS- and MetS+ subjects. The mRNA expression of 93 genes implicated in obesity-associated WAT dysfunction was analysed by RT-qPCR in both fat depots. MetS+ females showed higher adipocyte hypertrophy in both fat depots and increased fibrosis and expression of macrophage and hypoxia markers in SAT. Transcriptional data suggest increased fatty acid oxidation in SAT and impaired thermogenesis and extracellular matrix remodelling in VAT from MetS+ subjects. A sPLS-DA model, including SAT expression of PPARA and LEPR genes identified MetS with an AUC = 0.87. Despite equal age, BMI and body composition, MetS+ females display morphological and transcriptional differences in both WAT depots, especially in SAT. These factors may contribute to the transition to MetS.
Collapse
Affiliation(s)
- Óscar Osorio-Conles
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Correspondence: (Ó.O.-C.); (A.d.H.); Tel.: +34-932275707 (ext. 2910) (Ó.O.-C.); +34-932279846 (A.d.H.); Fax: +34932275589 (A.d.H.)
| | - Arturo Vega-Beyhart
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
| | - Ainitze Ibarzabal
- Gastrointestinal Surgery Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (A.I.); (J.M.B.)
| | - José María Balibrea
- Gastrointestinal Surgery Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (A.I.); (J.M.B.)
| | - Josep Vidal
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Ana de Hollanda
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Fisiopatologia de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Correspondence: (Ó.O.-C.); (A.d.H.); Tel.: +34-932275707 (ext. 2910) (Ó.O.-C.); +34-932279846 (A.d.H.); Fax: +34932275589 (A.d.H.)
| |
Collapse
|
40
|
Reversible lysine fatty acylation of an anchoring protein mediates adipocyte adrenergic signaling. Proc Natl Acad Sci U S A 2022; 119:2119678119. [PMID: 35149557 PMCID: PMC8851525 DOI: 10.1073/pnas.2119678119] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 01/05/2023] Open
Abstract
N-myristoylation on glycine is an irreversible modification that has long been recognized to govern protein localization and function. In contrast, the biological roles of lysine myristoylation remain ill-defined. We demonstrate that the cytoplasmic scaffolding protein, gravin-α/A kinase-anchoring protein 12, is myristoylated on two lysine residues embedded in its carboxyl-terminal protein kinase A (PKA) binding domain. Histone deacetylase 11 (HDAC11) docks to an adjacent region of gravin-α and demyristoylates these sites. In brown and white adipocytes, lysine myristoylation of gravin-α is required for signaling via β2- and β3-adrenergic receptors (β-ARs), which are G protein-coupled receptors (GPCRs). Lysine myristoylation of gravin-α drives β-ARs to lipid raft membrane microdomains, which results in PKA activation and downstream signaling that culminates in protective thermogenic gene expression. These findings define reversible lysine myristoylation as a mechanism for controlling GPCR signaling and highlight the potential of inhibiting HDAC11 to manipulate adipocyte phenotypes for therapeutic purposes.
Collapse
|
41
|
Abstract
The role of β-adrenergic receptors (βARs) in adipose tissue to promote lipolysis and the release of fatty acids and nonshivering thermogenesis in brown fat has been studied for so many decades that one would think there is nothing left to discover. With the rediscovery of brown fat in humans and renewed interest in UCP1 and uncoupled mitochondrial respiration, it seems that a review of adipose tissue as an organ, pivotal observations, and the investigators who made them would be instructive to understanding where the field stands now. The discovery of the β3-adrenergic receptor was important for accurately defining the pharmacology of the adipocyte, while the clinical targeting of this receptor for obesity and metabolic disease has had its highs and lows. Many questions still remain about how βARs regulate adipocyte metabolism and the signaling molecules through which they do it.
Collapse
Affiliation(s)
- Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA;
| |
Collapse
|
42
|
Shamsi F, Wang CH, Tseng YH. The evolving view of thermogenic adipocytes - ontogeny, niche and function. Nat Rev Endocrinol 2021; 17:726-744. [PMID: 34625737 PMCID: PMC8814904 DOI: 10.1038/s41574-021-00562-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 12/12/2022]
Abstract
The worldwide incidence of obesity and its sequelae, such as type 2 diabetes mellitus, have reached pandemic levels. Central to the development of these metabolic disorders is adipose tissue. White adipose tissue stores excess energy, whereas brown adipose tissue (BAT) and beige (also known as brite) adipose tissue dissipate energy to generate heat in a process known as thermogenesis. Strategies that activate and expand BAT and beige adipose tissue increase energy expenditure in animal models and offer therapeutic promise to treat obesity. A better understanding of the molecular mechanisms underlying the development of BAT and beige adipose tissue and the activation of thermogenic function is the key to creating practical therapeutic interventions for obesity and metabolic disorders. In this Review, we discuss the regulation of the tissue microenvironment (the adipose niche) and inter-organ communication between BAT and other tissues. We also cover the activation of BAT and beige adipose tissue in response to physiological cues (such as cold exposure, exercise and diet). We highlight advances in harnessing the therapeutic potential of BAT and beige adipose tissue by genetic, pharmacological and cell-based approaches in obesity and metabolic disorders.
Collapse
Affiliation(s)
- Farnaz Shamsi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA
| | - Chih-Hao Wang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
43
|
Rebello CJ, Coulter AA, Reaume AG, Cong W, Cusimano LA, Greenway FL. MLR-1023 Treatment in Mice and Humans Induces a Thermogenic Program, and Menthol Potentiates the Effect. Pharmaceuticals (Basel) 2021; 14:ph14111196. [PMID: 34832978 PMCID: PMC8625945 DOI: 10.3390/ph14111196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/09/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022] Open
Abstract
A glucose-lowering medication that acts by a different mechanism than metformin, or other approved diabetes medications, can supplement monotherapies when patients fail to meet blood glucose goals. We examined the actions underlying the effects of an insulin sensitizer, tolimidone (MLR-1023) and investigated its effects on body weight. Diet-induced obesity (CD1/ICR) and type 2 diabetes (db/db) mouse models were used to study the effect of MLR-1023 on metabolic outcomes and to explore its synergy with menthol. We also examined the efficacy of MLR-1023 alone in a clinical trial (NCT02317796), as well as in combination with menthol in human adipocytes. MLR-1023 produced weight loss in humans in four weeks, and in mice fed a high-fat diet it reduced weight gain and fat mass without affecting food intake. In human adipocytes from obese donors, the upregulation of Uncoupling Protein 1, Glucose (UCP)1, adiponectin, Glucose Transporter Type 4 (GLUT4), Adipose Triglyceride Lipase (ATGL), Carnitine palmitoyltransferase 1 beta (CPT1β), and Transient Receptor Potential Melastin (TRPM8) mRNA expression suggested the induction of thermogenesis. The TRPM8 agonist, menthol, potentiated the effect of MLR-1023 on the upregulation of genes for energy expenditure and insulin sensitivity in human adipocytes, and reduced fasting blood glucose in mice. The amplification of the thermogenic program by MLR-1023 and menthol in the absence of adrenergic activation will likely be well-tolerated, and bears investigation in a clinical trial.
Collapse
Affiliation(s)
- Candida J. Rebello
- Clinical Trials Unit, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; (C.J.R.); (A.A.C.)
| | - Ann A. Coulter
- Clinical Trials Unit, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; (C.J.R.); (A.A.C.)
| | - Andrew G. Reaume
- Melior Discovery Inc., 860 Springdale Drive, Exton, PA 19341, USA; (A.G.R.); (W.C.)
| | - Weina Cong
- Melior Discovery Inc., 860 Springdale Drive, Exton, PA 19341, USA; (A.G.R.); (W.C.)
| | - Luke A. Cusimano
- Cusimano Plastic and Reconstructive Surgery, 5233 Dijon Dr, Baton Rouge, LA 70808, USA;
| | - Frank L. Greenway
- Clinical Trials Unit, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; (C.J.R.); (A.A.C.)
- Correspondence: ; Tel.: +1-(225)-763-2576; Fax: +1-(225)-763-3022
| |
Collapse
|
44
|
Human Brown Adipose Tissue and Metabolic Health: Potential for Therapeutic Avenues. Cells 2021; 10:cells10113030. [PMID: 34831253 PMCID: PMC8616549 DOI: 10.3390/cells10113030] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/31/2022] Open
Abstract
Obesity-associated metabolic abnormalities comprise a cluster of conditions including dyslipidemia, insulin resistance, diabetes and cardiovascular diseases that has affected more than 650 million people all over the globe. Obesity results from the accumulation of white adipose tissues mainly due to the chronic imbalance of energy intake and energy expenditure. A variety of approaches to treat or prevent obesity, including lifestyle interventions, surgical weight loss procedures and pharmacological approaches to reduce energy intake and increase energy expenditure have failed to substantially decrease the prevalence of obesity. Brown adipose tissue (BAT), the primary source of thermogenesis in infants and small mammals may represent a promising therapeutic target to treat obesity by promoting energy expenditure through non-shivering thermogenesis mediated by mitochondrial uncoupling protein 1 (UCP1). Since the confirmation of functional BAT in adult humans by several groups, approximately a decade ago, and its association with a favorable metabolic phenotype, intense interest on the significance of BAT in adult human physiology and metabolic health has emerged within the scientific community to explore its therapeutic potential for the treatment of obesity and metabolic diseases. A substantially decreased BAT activity in individuals with obesity indicates a role for BAT in the setting of human obesity. On the other hand, BAT mass and its prevalence correlate with lower body mass index (BMI), decreased age and lower glucose levels, leading to a lower incidence of cardio-metabolic diseases. The increased cold exposure in adult humans with undetectable BAT was associated with decreased body fat mass and increased insulin sensitivity. A deeper understanding of the role of BAT in human metabolic health and its interrelationship with body fat distribution and deciphering proper strategies to increase energy expenditure, by either increasing functional BAT mass or inducing white adipose browning, holds the promise for possible therapeutic avenues for the treatment of obesity and associated metabolic disorders.
Collapse
|
45
|
Greenfield AM, Charkoudian N, Alba BK. Influences of ovarian hormones on physiological responses to cold in women. Temperature (Austin) 2021; 9:23-45. [DOI: 10.1080/23328940.2021.1953688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Andrew Martin Greenfield
- Thermal & Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
- Oak Ridge Institute of Science and Education, Belcamp, MD, USA
| | - Nisha Charkoudian
- Thermal & Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Billie Katherine Alba
- Thermal & Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| |
Collapse
|
46
|
Cero C, Lea HJ, Zhu KY, Shamsi F, Tseng YH, Cypess AM. β3-Adrenergic receptors regulate human brown/beige adipocyte lipolysis and thermogenesis. JCI Insight 2021; 6:e139160. [PMID: 34100382 PMCID: PMC8262278 DOI: 10.1172/jci.insight.139160] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
β3-Adrenergic receptors (β3-ARs) are the predominant regulators of rodent brown adipose tissue (BAT) thermogenesis. However, in humans, the physiological relevance of BAT and β3-AR remains controversial. Herein, using primary human adipocytes from supraclavicular neck fat and immortalized brown/beige adipocytes from deep neck fat from 2 subjects, we demonstrate that the β3-AR plays a critical role in regulating lipolysis, glycolysis, and thermogenesis. Silencing of the β3-AR compromised genes essential for thermogenesis, fatty acid metabolism, and mitochondrial mass. Functionally, reduction of β3-AR lowered agonist-mediated increases in intracellular cAMP, lipolysis, and lipolysis-activated, uncoupling protein 1-mediated thermogenic capacity. Furthermore, mirabegron, a selective human β3-AR agonist, stimulated BAT lipolysis and thermogenesis, and both processes were lost after silencing β3-AR expression. This study highlights that β3-ARs in human brown/beige adipocytes are required to maintain multiple components of the lipolytic and thermogenic cellular machinery and that β3-AR agonists could be used to achieve metabolic benefit in humans.
Collapse
Affiliation(s)
- Cheryl Cero
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Hannah J Lea
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Kenneth Y Zhu
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Farnaz Shamsi
- Integrative Physiology and Metabolism Section, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Yu-Hua Tseng
- Integrative Physiology and Metabolism Section, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| |
Collapse
|
47
|
Suchacki KJ, Stimson RH. Nutritional Regulation of Human Brown Adipose Tissue. Nutrients 2021; 13:nu13061748. [PMID: 34063868 PMCID: PMC8224032 DOI: 10.3390/nu13061748] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
The recent identification of brown adipose tissue in adult humans offers a new strategy to increase energy expenditure to treat obesity and associated metabolic disease. While white adipose tissue (WAT) is primarily for energy storage, brown adipose tissue (BAT) is a thermogenic organ that increases energy expenditure to generate heat. BAT is activated upon cold exposure and improves insulin sensitivity and lipid clearance, highlighting its beneficial role in metabolic health in humans. This review provides an overview of BAT physiology in conditions of overnutrition (obesity and associated metabolic disease), undernutrition and in conditions of altered fat distribution such as lipodystrophy. We review the impact of exercise, dietary macronutrients and bioactive compounds on BAT activity. Finally, we discuss the therapeutic potential of dietary manipulations or supplementation to increase energy expenditure and BAT thermogenesis. We conclude that chronic nutritional interventions may represent a useful nonpharmacological means to enhance BAT mass and activity to aid weight loss and/or improve metabolic health.
Collapse
|
48
|
Targeting Energy Expenditure-Drugs for Obesity Treatment. Pharmaceuticals (Basel) 2021; 14:ph14050435. [PMID: 34066399 PMCID: PMC8148206 DOI: 10.3390/ph14050435] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/18/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
Obesity and overweight are associated with lethal diseases. In this context, obese and overweight individuals infected by COVID-19 are at greater risk of dying. Obesity is treated by three main pharmaceutical approaches, namely suppressing appetite, reducing energy intake by impairing absorption, and increasing energy expenditure. Most compounds used for the latter were first envisaged for other medical uses. However, several candidates are now being developed explicitly for targeting obesity by increasing energy expenditure. This review analyzes the compounds that show anti-obesity activity exerted through the energy expenditure pathway. They are classified on the basis of their development status: FDA-approved, Withdrawn, Clinical Trials, and Under Development. The chemical nature, target, mechanisms of action, and description of the current stage of development are described for each one.
Collapse
|
49
|
McNeill BT, Suchacki KJ, Stimson RH. MECHANISMS IN ENDOCRINOLOGY: Human brown adipose tissue as a therapeutic target: warming up or cooling down? Eur J Endocrinol 2021; 184:R243-R259. [PMID: 33729178 PMCID: PMC8111330 DOI: 10.1530/eje-20-1439] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
Excessive accumulation of white adipose tissue leads to obesity and its associated metabolic health consequences such as type 2 diabetes and cardiovascular disease. Several approaches to treat or prevent obesity including public health interventions, surgical weight loss, and pharmacological approaches to reduce caloric intake have failed to substantially modify the increasing prevalence of obesity. The (re-)discovery of active brown adipose tissue (BAT) in adult humans approximately 15 years ago led to a resurgence in research into whether BAT activation could be a novel therapy for the treatment of obesity. Upon cold stimulus, BAT activates and generates heat to maintain body temperature, thus increasing energy expenditure. Activation of BAT may provide a unique opportunity to increase energy expenditure without the need for exercise. However, much of the underlying mechanisms surrounding BAT activation are still being elucidated and the effectiveness of BAT as a therapeutic target has not been realised. Research is ongoing to determine how best to expand BAT mass and activate existing BAT; approaches include cold exposure, pharmacological stimulation using sympathomimetics, browning agents that induce formation of thermogenic beige adipocytes in white adipose depots, and the identification of factors secreted by BAT with therapeutic potential. In this review, we discuss the caloric capacity and other metabolic benefits from BAT activation in humans and the role of metabolic tissues such as skeletal muscle in increasing energy expenditure. We discuss the potential of current approaches and the challenges of BAT activation as a novel strategy to treat obesity and metabolic disorders.
Collapse
Affiliation(s)
- Ben T McNeill
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, UK
| | - Karla J Suchacki
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, UK
| | - Roland H Stimson
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, UK
- Correspondence should be addressed to R H Stimson Email
| |
Collapse
|
50
|
Finlin BS, Memetimin H, Zhu B, Confides AL, Vekaria HJ, El Khouli RH, Johnson ZR, Westgate PM, Chen J, Morris AJ, Sullivan PG, Dupont-Versteegden EE, Kern PA. Pioglitazone does not synergize with mirabegron to increase beige fat or further improve glucose metabolism. JCI Insight 2021; 6:143650. [PMID: 33571166 PMCID: PMC8026187 DOI: 10.1172/jci.insight.143650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Beige and brown adipose tissue (BAT) are associated with improved metabolic homeostasis. We recently reported that the β3-adrenergic receptor agonist mirabegron induced beige adipose tissue in obese insulin-resistant subjects, and this was accompanied by improved glucose metabolism. Here we evaluated pioglitazone treatment with a combination pioglitazone and mirabegron treatment and compared these with previously published data evaluating mirabegron treatment alone. Both drugs were used at FDA-approved dosages. METHODS We measured BAT by PET CT scans, measured beige adipose tissue by immunohistochemistry, and comprehensively characterized glucose and lipid homeostasis and insulin sensitivity by euglycemic clamp and oral glucose tolerance tests. Subcutaneous white adipose tissue, muscle fiber type composition and capillary density, lipotoxicity, and systemic inflammation were evaluated by immunohistochemistry, gene expression profiling, mass spectroscopy, and ELISAs. RESULTS Treatment with pioglitazone or the combination of pioglitazone and mirabegron increased beige adipose tissue protein marker expression and improved insulin sensitivity and glucose homeostasis, but neither treatment induced BAT in these obese subjects. When the magnitude of the responses to the treatments was evaluated, mirabegron was found to be the most effective at inducing beige adipose tissue. Although monotherapy with either mirabegron or pioglitazone induced adipose beiging, combination treatment resulted in less beiging than either alone. The 3 treatments also had different effects on muscle fiber type switching and capillary density. CONCLUSION The addition of pioglitazone to mirabegron treatment does not enhance beiging or increase BAT in obese insulin-resistant research participants. TRIAL REGISTRATION ClinicalTrials.gov NCT02919176. FUNDING NIH DK112282 and P20GM103527 and Clinical and Translational Science Awards grant UL1TR001998.
Collapse
Affiliation(s)
- Brian S Finlin
- Division of Endocrinology, Department of Internal Medicine, College of Medicine.,Barnstable Brown Diabetes and Obesity Center
| | - Hasiyet Memetimin
- Division of Endocrinology, Department of Internal Medicine, College of Medicine.,Barnstable Brown Diabetes and Obesity Center
| | - Beibei Zhu
- Division of Endocrinology, Department of Internal Medicine, College of Medicine.,Barnstable Brown Diabetes and Obesity Center
| | - Amy L Confides
- Department of Physical Therapy and Center for Muscle Biology, College of Health Sciences
| | | | | | - Zachary R Johnson
- Division of Endocrinology, Department of Internal Medicine, College of Medicine
| | | | - Jianzhong Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, Kentucky, USA.,Lexington Veterans Affairs Medical Center, Lexington, Kentucky, USA
| | - Andrew J Morris
- Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, Kentucky, USA.,Lexington Veterans Affairs Medical Center, Lexington, Kentucky, USA
| | | | | | - Philip A Kern
- Division of Endocrinology, Department of Internal Medicine, College of Medicine.,Barnstable Brown Diabetes and Obesity Center
| |
Collapse
|