1
|
Erdil E, Becker AS, Schwyzer M, Martinez-Tellez B, Ruiz JR, Sartoretti T, Vargas HA, Burger AI, Chirindel A, Wild D, Zamboni N, Deplancke B, Gardeux V, Maushart CI, Betz MJ, Wolfrum C, Konukoglu E. Predicting standardized uptake value of brown adipose tissue from CT scans using convolutional neural networks. Nat Commun 2024; 15:8402. [PMID: 39333526 PMCID: PMC11436835 DOI: 10.1038/s41467-024-52622-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 09/13/2024] [Indexed: 09/29/2024] Open
Abstract
The standard method for identifying active Brown Adipose Tissue (BAT) is [18F]-Fluorodeoxyglucose ([18F]-FDG) PET/CT imaging, which is costly and exposes patients to radiation, making it impractical for population studies. These issues can be addressed with computational methods that predict [18F]-FDG uptake by BAT from CT; earlier population studies pave the way for developing such methods by showing some correlation between the Hounsfield Unit (HU) of BAT in CT and the corresponding [18F]-FDG uptake in PET. In this study, we propose training convolutional neural networks (CNNs) to predict [18F]-FDG uptake by BAT from unenhanced CT scans in the restricted regions that are likely to contain BAT. Using the Attention U-Net architecture, we perform experiments on datasets from four different cohorts, the largest study to date. We segment BAT regions using predicted [18F]-FDG uptake values, achieving 23% to 40% better accuracy than conventional CT thresholding. Additionally, BAT volumes computed from the segmentations distinguish the subjects with and without active BAT with an AUC of 0.8, compared to 0.6 for CT thresholding. These findings suggest CNNs can facilitate large-scale imaging studies more efficiently and cost-effectively using only CT.
Collapse
Affiliation(s)
- Ertunc Erdil
- Computer Vision Lab., ETH Zurich, Zurich, Switzerland.
| | - Anton S Becker
- Computer Vision Lab., ETH Zurich, Zurich, Switzerland
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Institute for Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Moritz Schwyzer
- Institute for Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Borja Martinez-Tellez
- Department of Nursing, Physiotherapy and Medicine and SPORT Research Group (CTS-1024), CERNEP Research Center, University of Almería, Almería, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Granada, Spain
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jonatan R Ruiz
- Department of Physical Education and Sports, Faculty of Sports Science, Sport and Health University Research Institute (iMUDS), University of Granada, 18071, Granada, Spain
- Instituto de Investigación Biosanitaria, Ibs.Granada, Granada, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Thomas Sartoretti
- Institute for Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - H Alberto Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Irene Burger
- Department of Nuclear Medicine, University Zurich Hospital, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Alin Chirindel
- Department of Radiology and Nuclear Medicine, University Hospital of Basel, Basel, Switzerland
| | - Damian Wild
- Department of Radiology and Nuclear Medicine, University Hospital of Basel, Basel, Switzerland
| | - Nicola Zamboni
- Swiss Multi-Omics Center, ETH Zürich, Zürich, Switzerland
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | - Vincent Gardeux
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | - Claudia Irene Maushart
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Matthias Johannes Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Christian Wolfrum
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Ender Konukoglu
- Computer Vision Lab., ETH Zurich, Zurich, Switzerland
- The LOOP Zürich - Medical Research Center, Zürich, Switzerland
| |
Collapse
|
2
|
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
|
3
|
Cai Z, Zhong Q, Feng Y, Wang Q, Zhang Z, Wei C, Yin Z, Liang C, Liew CW, Kazak L, Cypess AM, Liu Z, Cai K. Non-invasive mapping of brown adipose tissue activity with magnetic resonance imaging. Nat Metab 2024; 6:1367-1379. [PMID: 39054361 PMCID: PMC11272596 DOI: 10.1038/s42255-024-01082-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 06/14/2024] [Indexed: 07/27/2024]
Abstract
Thermogenic brown adipose tissue (BAT) has a positive impact on whole-body metabolism. However, in vivo mapping of BAT activity typically relies on techniques involving ionizing radiation, such as [18F]fluorodeoxyglucose ([18F]FDG) positron emission tomography (PET) and computed tomography (CT). Here we report a noninvasive metabolic magnetic resonance imaging (MRI) approach based on creatine chemical exchange saturation transfer (Cr-CEST) contrast to assess in vivo BAT activity in rodents and humans. In male rats, a single dose of the β3-adrenoceptor agonist (CL 316,243) or norepinephrine, as well as cold exposure, triggered a robust elevation of the Cr-CEST MRI signal, which was consistent with the [18F]FDG PET and CT data and 1H nuclear magnetic resonance measurements of creatine concentration in BAT. We further show that Cr-CEST MRI detects cold-stimulated BAT activation in humans (both males and females) using a 3T clinical scanner, with data-matching results from [18F]FDG PET and CT measurements. This study establishes Cr-CEST MRI as a promising noninvasive and radiation-free approach for in vivo mapping of BAT activity.
Collapse
Affiliation(s)
- Zimeng Cai
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Qiaoling Zhong
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Image Processing & Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence & Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China
| | - Qian Wang
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Zuoman Zhang
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cailv Wei
- School of Medicine, Shenzhen Campus, Sun Yat-sen University, Shenzhen, China
| | - Zhinan Yin
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Chong Wee Liew
- Physiology and Biophysics Department, University of Illinois at Chicago, Chicago, IL, USA
| | - Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China.
| | - Kejia Cai
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
4
|
Carpentier AC, Blondin DP. Is stimulation of browning of human adipose tissue a relevant therapeutic target? ANNALES D'ENDOCRINOLOGIE 2024; 85:184-189. [PMID: 38871497 DOI: 10.1016/j.ando.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Brown adipose tissue (BAT) and beige adipose tissues are important contributors to cold-induced whole body thermogenesis in rodents. The documentation in humans of cold- and ß-adrenergic receptor agonist-stimulated BAT glucose uptake using positron emission tomography (PET) and of a decrease of this response in individuals with cardiometabolic disorders led to the suggestion that BAT/beige adipose tissues could be relevant targets for prevention and treatment of these conditions. In this brief review, we will critically assess this question by first describing the basic rationale for this affirmation, second by examining the evidence in human studies, and third by discussing the possible means to activate the thermogenic response of these tissues in humans.
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, Sherbrooke, Québec, Canada.
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
5
|
Shi L, Yang J, Tao Z, Zheng L, Bui T, Alonso R, Yue F, Cheng Z. Loss of FoxO1 activates an alternate mechanism of mitochondrial quality control for healthy adipose browning. Clin Sci (Lond) 2024; 138:371-385. [PMID: 38469619 PMCID: PMC10932742 DOI: 10.1042/cs20230973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/13/2024]
Abstract
Browning of white adipose tissue is hallmarked by increased mitochondrial density and metabolic improvements. However, it remains largely unknown how mitochondrial turnover and quality control are regulated during adipose browning. In the present study, we found that mice lacking adipocyte FoxO1, a transcription factor that regulates autophagy, adopted an alternate mechanism of mitophagy to maintain mitochondrial turnover and quality control during adipose browning. Post-developmental deletion of adipocyte FoxO1 (adO1KO) suppressed Bnip3 but activated Fundc1/Drp1/OPA1 cascade, concurrent with up-regulation of Atg7 and CTSL. In addition, mitochondrial biogenesis was stimulated via the Pgc1α/Tfam pathway in adO1KO mice. These changes were associated with enhanced mitochondrial homeostasis and metabolic health (e.g., improved glucose tolerance and insulin sensitivity). By contrast, silencing Fundc1 or Pgc1α reversed the changes induced by silencing FoxO1, which impaired mitochondrial quality control and function. Ablation of Atg7 suppressed mitochondrial turnover and function, causing metabolic disorder (e.g., impaired glucose tolerance and insulin sensitivity), regardless of elevated markers of adipose browning. Consistently, suppression of autophagy via CTSL by high-fat diet was associated with a reversal of adO1KO-induced benefits. Our data reveal a unique role of FoxO1 in coordinating mitophagy receptors (Bnip3 and Fundc1) for a fine-tuned mitochondrial turnover and quality control, underscoring autophagic clearance of mitochondria as a prerequisite for healthy browning of adipose tissue.
Collapse
Affiliation(s)
- Limin Shi
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
- Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611, U.S.A
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32610, U.S.A
| | - Jinying Yang
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
- Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611, U.S.A
| | - Zhipeng Tao
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, U.S.A
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, U.S.A
| | - Louise Zheng
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, U.S.A
| | - Tyler F. Bui
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
| | - Ramon L. Alonso
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
| | - Feng Yue
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, U.S.A
| | - Zhiyong Cheng
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
- Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611, U.S.A
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32610, U.S.A
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, U.S.A
| |
Collapse
|
6
|
Jalloul W, Moscalu M, Moscalu R, Jalloul D, Grierosu IC, Gutu M, Haba D, Mocanu V, Gutu MM, Stefanescu C. Are MTV and TLG Accurate for Quantifying the Intensity of Brown Adipose Tissue Activation? Biomedicines 2024; 12:151. [PMID: 38255256 PMCID: PMC10813038 DOI: 10.3390/biomedicines12010151] [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: 12/20/2023] [Revised: 12/31/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Recent research has suggested that one novel mechanism of action for anti-obesity medications is to stimulate the activation of brown adipose tissue (BAT). 18FDG PET/CT remains the gold standard for defining and quantifying BAT. SUVmax is the most often used quantification tool in clinical practice. However, this parameter does not reflect the entire BAT volume. As a potential method for precisely evaluating BAT, we have utilised metabolic tumour volume (MTV) and total lesion glycolysis (TLG) to answer the question: Are MTV and TLG accurate in quantifying the intensity of BAT activation? After analysing the total number of oncological 18F-FDG PET/CT scans between 2021-2023, we selected patients with active BAT. Based on the BAT SUVmax, the patients were divided into BAT-moderate activation (MA) vs. BAT-high activation (HA). Furthermore, we statistically analysed the accuracy of TLG and MTV in assessing BAT activation intensity. The results showed that both parameters increased their predictive value regarding BAT activation, and presented a significantly high sensitivity and specificity for the correct classification of BAT activation intensity. To conclude, these parameters could be important indicators with increased accuracy for classifying BAT expression, and could bring additional information about the volume of BAT to complement the limitations of the SUVmax.
Collapse
Affiliation(s)
- Wael Jalloul
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (W.J.); (D.J.); (I.C.G.); (C.S.)
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Roxana Moscalu
- Manchester Academic Health Science Centre, Cell Matrix Biology and Regenerative Medicine, The University of Manchester, Manchester M13 9PT, UK;
| | - Despina Jalloul
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (W.J.); (D.J.); (I.C.G.); (C.S.)
| | - Irena Cristina Grierosu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (W.J.); (D.J.); (I.C.G.); (C.S.)
| | - Mihaela Gutu
- County Hospital of Emergency “Saint John the New”, 720224 Suceava, Romania; (M.G.); (M.M.G.)
| | - Danisia Haba
- Department 1 Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Veronica Mocanu
- Department of Morpho-Functional Sciences (Pathophysiology), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Mihai Marius Gutu
- County Hospital of Emergency “Saint John the New”, 720224 Suceava, Romania; (M.G.); (M.M.G.)
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (W.J.); (D.J.); (I.C.G.); (C.S.)
| |
Collapse
|
7
|
McClave SA, Martindale RG. Browning of white adipose tissue may be an appropriate adaptive response to critical illness. JPEN J Parenter Enteral Nutr 2024; 48:37-45. [PMID: 37908064 DOI: 10.1002/jpen.2576] [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: 07/25/2023] [Revised: 10/16/2023] [Accepted: 10/28/2023] [Indexed: 11/02/2023]
Abstract
Both the baseline amount of brown adipose tissue (BAT) and the capacity to stimulate browning of white adipose tissue (WAT) may provide a protective effect to the patient in a critical care setting. Critical illness is associated with reduced mitochondrial volume and function resulting in the increased production of reactive oxygen species, greater demand for adenosine triphosphate, a switch to uncoupled fat metabolism, and hibernation of the organelle, which all contribute to multiple organ failure. Increasing insulin resistance, decreasing fatty acid oxidation, and dependence on carbohydrate metabolism result. Browning of WAT may oppose many of these adverse effects. The presence of BAT and the changes associated with browning may help dissipate oxidative stress, increase consumption and utilization of metabolites, and reduce pro-inflammatory actions. The number of mitochondria increases, and there is greater infiltration of macrophages into adipose tissue. A shift occurs in macrophage expression from the M1 to M2 phenotype, an effect which further dampens inflammation, increases insulin sensitivity, and improves tissue healing and remodeling. Any benefit from these responses may be lost in the disease states of chronic hypermetabolism (such as burns or cancer cachexia) in which the persistence of these physiologic effects may become detrimental, contributing to excessive weight loss, adipose wasting, and loss of lean body mass. This paper discusses the plasticity of adipose tissue and whether shifts in its physiology provide clinical advantages in the intensive care unit.
Collapse
Affiliation(s)
- Stephen A McClave
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Robert G Martindale
- Department of Surgery, Oregon Health Sciences University, Portland, Oregon, USA
| |
Collapse
|
8
|
Zhu Y, Liu W, Qi Z. Adipose tissue browning and thermogenesis under physiologically energetic challenges: a remodelled thermogenic system. J Physiol 2024; 602:23-48. [PMID: 38019069 DOI: 10.1113/jp285269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/16/2023] [Indexed: 11/30/2023] Open
Abstract
Metabolic diseases such as obesity and diabetes are often thought to be caused by reduced energy expenditure, which poses a serious threat to human health. Cold exposure, exercise and caloric restriction have been shown to promote adipose tissue browning and thermogenesis. These physiological interventions increase energy expenditure and thus have emerged as promising strategies for mitigating metabolic disorders. However, that increased adipose tissue browning and thermogenesis elevate thermogenic consumption is not a reasonable explanation when humans and animals confront energetic challenges imposed by these interventions. In this review, we collected numerous results on adipose tissue browning and whitening and evaluated this bi-directional conversion of adipocytes from the perspective of energy homeostasis. Here, we propose a new interpretation of the role of adipose tissue browning under energetic challenges: increased adipose tissue browning and thermogenesis under energy challenge is not to enhance energy expenditure, but to reestablish a more economical thermogenic pattern to maintain the core body temperature. This can be achieved by enhancing the contribution of non-shivering thermogenesis (adipose tissue browning and thermogenesis) and lowering shivering thermogenesis and high intensity shivering. Consequently, the proportion of heat production in fat increases and that in skeletal muscle decreases, enabling skeletal muscle to devote more energy reserves to overcoming environmental stress.
Collapse
Affiliation(s)
- Yupeng Zhu
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, China
- School of Physical Education and Health, East China Normal University, Shanghai, China
- Sino-French Joint Research Center of Sport Science, East China Normal University, Shanghai, China
| | - Weina Liu
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, China
- School of Physical Education and Health, East China Normal University, Shanghai, China
| | - Zhengtang Qi
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, China
- School of Physical Education and Health, East China Normal University, Shanghai, China
| |
Collapse
|
9
|
Ziqubu K, Dludla PV, Mabhida SE, Jack BU, Keipert S, Jastroch M, Mazibuko-Mbeje SE. Brown adipose tissue-derived metabolites and their role in regulating metabolism. Metabolism 2024; 150:155709. [PMID: 37866810 DOI: 10.1016/j.metabol.2023.155709] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/28/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
The discovery and rejuvenation of metabolically active brown adipose tissue (BAT) in adult humans have offered a new approach to treat obesity and metabolic diseases. Beyond its accomplished role in adaptive thermogenesis, BAT secretes signaling molecules known as "batokines", which are instrumental in regulating whole-body metabolism via autocrine, paracrine, and endocrine action. In addition to the intrinsic BAT metabolite-oxidizing activity, the endocrine functions of these molecules may help to explain the association between BAT activity and a healthy systemic metabolic profile. Herein, we review the evidence that underscores the significance of BAT-derived metabolites, especially highlighting their role in controlling physiological and metabolic processes involving thermogenesis, substrate metabolism, and other essential biological processes. The conversation extends to their capacity to enhance energy expenditure and mitigate features of obesity and its related metabolic complications. Thus, metabolites derived from BAT may provide new avenues for the discovery of metabolic health-promoting drugs with far-reaching impacts. This review aims to dissect the complexities of the secretory role of BAT in modulating local and systemic metabolism in metabolic health and disease.
Collapse
Affiliation(s)
- Khanyisani Ziqubu
- Department of Biochemistry, North-West University, Mmabatho 2745, South Africa
| | - Phiwayinkosi V Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Sihle E Mabhida
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Babalwa U Jack
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Susanne Keipert
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | | |
Collapse
|
10
|
Neri D, Ramos-Lobo AM, Lee S, Lafond A, Zeltser LM. Rearing mice at 22°C programs increased capacity to respond to chronic exposure to cold but not high fat diet. Mol Metab 2023; 73:101740. [PMID: 37211277 PMCID: PMC10248272 DOI: 10.1016/j.molmet.2023.101740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023] Open
Abstract
OBJECTIVE Rodent models raised at environmental temperatures of 21-22 °C are increasingly switched to thermoneutral housing conditions in adulthood to better capture human physiology. We quantified the developmental effects of rearing mice at an ambient temperature of 22 °C vs. 30 °C on metabolic responses to cold and high fat diet (HFD) in adulthood. METHODS Mice were reared from birth to 8 weeks of age at 22 °C or 30 °C, when they were acclimated to single housing at the same temperature for 2-3 weeks in indirect calorimetry cages. Energy expenditure attributable to basal metabolic rate, physical activity, thermic effect of food, and adaptive cold- or diet-induced thermogenesis was calculated. Responses to cooling were evaluated by decreasing the ambient temperature from 22 °C to 14 °C, while responses to HFD feeding were assessed at 30 °C. Influences of rearing temperature on thermogenic responses that emerge over hours, days and weeks were assessed by maintaining mice in the indirect calorimetry cages throughout the study. RESULTS At an ambient temperature of 22 °C, total energy expenditure (TEE) was 12-16% higher in mice reared at 22 °C as compared to 30 °C. Rearing temperature had no effect on responses in the first hours or week of the 14 °C challenge. Differences emerged in the third week, when TEE increased an additional 10% in mice reared at 22 °C, but mice reared at 30 °C could not sustain this level of cold-induced thermogenesis. Rearing temperature only affected responses to HFD during the first week, due to differences in the timing but not the strength of metabolic adaptations. CONCLUSION Rearing at 22 °C does not have a lasting effect on metabolic adaptations to HFD at thermoneutrality, but it programs an enhanced capacity to respond to chronic cold challenges in adulthood. These findings highlight the need to consider rearing temperature when using mice to model cold-induced thermogenesis.
Collapse
Affiliation(s)
- Daniele Neri
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Angela M Ramos-Lobo
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Seoeun Lee
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alexandre Lafond
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lori M Zeltser
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
11
|
Blondin DP. Human thermogenic adipose tissue. Curr Opin Genet Dev 2023; 80:102054. [PMID: 37269791 DOI: 10.1016/j.gde.2023.102054] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/11/2023] [Accepted: 04/28/2023] [Indexed: 06/05/2023]
Abstract
Human thermogenic adipose tissue has long been touted as a promising therapeutic target for obesity and its associated metabolic diseases. Here, we provide a brief overview of the current knowledge of in vivo human thermogenic adipose tissue metabolism. We explore the evidence provided by retrospective and prospective studies describing the association of brown adipose tissue (BAT) [18F]fluorodeoxyglucose accumulation and various cardiometabolic risk factors. Although these studies have been invaluable in generating hypothesis, it has also raised some questions about the reliability of this method as an indicator of BAT thermogenic capacity. We discuss the evidence in support of human BAT functioning as a local thermogenic organ and energy sink, as an endocrine organ, and as a biomarker of adipose tissue health.
Collapse
Affiliation(s)
- Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, 3001, 12th Ave North, Sherbrooke, Quebec J1H 5N4, Canada.
| |
Collapse
|
12
|
Shi L, Tao Z, Zheng L, Yang J, Hu X, Scott K, de Kloet A, Krause E, Collins JF, Cheng Z. FoxO1 regulates adipose transdifferentiation and iron influx by mediating Tgfβ1 signaling pathway. Redox Biol 2023; 63:102727. [PMID: 37156218 DOI: 10.1016/j.redox.2023.102727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 05/10/2023] Open
Abstract
Adipose plasticity is critical for metabolic homeostasis. Adipocyte transdifferentiation plays an important role in adipose plasticity, but the molecular mechanism of transdifferentiation remains incompletely understood. Here we show that the transcription factor FoxO1 regulates adipose transdifferentiation by mediating Tgfβ1 signaling pathway. Tgfβ1 treatment induced whitening phenotype in beige adipocytes, reducing UCP1 and mitochondrial capacity and enlarging lipid droplets. Deletion of adipose FoxO1 (adO1KO) dampened Tgfβ1 signaling by downregulating Tgfbr2 and Smad3 and induced browning of adipose tissue in mice, increasing UCP1 and mitochondrial content and activating metabolic pathways. Silencing FoxO1 also abolished the whitening effect of Tgfβ1 on beige adipocytes. The adO1KO mice exhibited a significantly higher energy expenditure, lower fat mass, and smaller adipocytes than the control mice. The browning phenotype in adO1KO mice was associated with an increased iron content in adipose tissue, concurrent with upregulation of proteins that facilitate iron uptake (DMT1 and TfR1) and iron import into mitochondria (Mfrn1). Analysis of hepatic and serum iron along with hepatic iron-regulatory proteins (ferritin and ferroportin) in the adO1KO mice revealed an adipose tissue-liver crosstalk that meets the increased iron requirement for adipose browning. The FoxO1-Tgfβ1 signaling cascade also underlay adipose browning induced by β3-AR agonist CL316243. Our study provides the first evidence of a FoxO1-Tgfβ1 axis in the regulation of adipose browning-whitening transdifferentiation and iron influx, which sheds light on the compromised adipose plasticity in conditions of dysregulated FoxO1 and Tgfβ1 signaling.
Collapse
Affiliation(s)
- Limin Shi
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA; Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA
| | - Zhipeng Tao
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, 24061, USA; Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Louise Zheng
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Jinying Yang
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA
| | - Xinran Hu
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA
| | - Karen Scott
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL32610, USA
| | - Annette de Kloet
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Eric Krause
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL32610, USA
| | - James F Collins
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA
| | - Zhiyong Cheng
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA; Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
13
|
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
|
14
|
Lv Y, Lv X, Feng J, Cheng F, Yu Z, Guan F, Chen L. (20R)-panaxadiol improves obesity by promoting white fat beigeing. Front Pharmacol 2023; 14:1071516. [PMID: 36909162 PMCID: PMC9992182 DOI: 10.3389/fphar.2023.1071516] [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: 10/16/2022] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Introduction: Obesity is an important cause of a range of metabolic diseases. However, the complex mechanisms of obesity and its related diseases make some weight loss methods ineffective or have safety issues. Ginseng, a specialty of Jilin Province in China with both edible and medicinal value, contains mainly ginsenosides and other components. In order to study the anti-obesity effect of ginseng, network pharmacology was used to predict and screen the active ingredients, action targets and signaling pathways of ginseng. We found (20R)-panaxadiol (PD) is a more desirable active ingredient due to its high drug-like properties and high bioavailability. Moreover, it is closely related to cAMP pathway which is more important in metabolism regulation. The corresponding pharmacodynamic targets of PD include ADRB2 (the gene encoding the β2-adrenoceptor receptor). Our study aimed to investigate whether Panaxadiol can promote white adipocyte beigeing and increase thermogenesis through modulating the β2/cAMP pathway to exert anti-obesity effects. Methods: In vivo, we established high-fat feeding obesity model, genotypically obese mice (ob/ob) model, and administered PD (10 mg/kg). PD treatment in ob/ob mice along with β2 receptor inhibitor ICI118551. In vitro, differentiated mature 3T3-L1 cells were given palmitate (PA) to induce hypertrophy model along with PD (20 μM). Results: The results of this study demonstrated that PD significantly reduced body weight, improved glucose tolerance and lipid levels in high-fat-induced obese mice and ob/ob mice, and also reduced lipid droplet size in PA-treated hypertrophic adipocytes in vitro. Molecular biology assays confirmed that cAMP response element binding protein (CREB) phosphorylation was increased after PD administration, and the expression of thermogenesis-related proteins UCP1, PRDM16 and mitochondrial biosynthesis-related proteins PGC-1α, TFAM and NRF1 were increased. Molecular docking results showed a low binding energy between β2 receptors and PD, indicating an affinity between the β2 receptor and PD. In addition, the β2 receptor inhibition, reversed the anti-obesity effect of PD on the body weight, lipid droplets, the expression of thermogenesis-related proteins and CREB phosphorylation in ob/ob mice. Discussion: These results suggest that PD may promote the expression of thermogenic proteins through phosphorylation of CREB via β2 receptor activation, and thus exert anti-obesity effects.
Collapse
Affiliation(s)
- Yuqian Lv
- Department of Pharmacology, School of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaoyan Lv
- Department of Clinical Laboratory, The Second Clinical Hospital Affiliated to Jilin University, Changchun, China
| | - Jianshu Feng
- Department of Pharmacology, School of Basic Medical Sciences, Jilin University, Changchun, China
| | - Fanghui Cheng
- Department of Pharmacology, School of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zhiyi Yu
- Department of Pharmacology, School of Basic Medical Sciences, Jilin University, Changchun, China.,Department of Physiology and Pharmacology, Department of Basic Medicine, Changchun Medical College, Changchun, China
| | - Fengying Guan
- Department of Pharmacology, School of Basic Medical Sciences, Jilin University, Changchun, China
| | - Li Chen
- Department of Pharmacology, School of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
15
|
Armani A, Feraco A, Camajani E, Gorini S, Lombardo M, Caprio M. Nutraceuticals in Brown Adipose Tissue Activation. Cells 2022; 11:cells11243996. [PMID: 36552762 PMCID: PMC9776638 DOI: 10.3390/cells11243996] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Obesity and its associated comorbidities have become pandemic, and challenge the global healthcare system. Lifestyle changes, nutritional interventions and phamaceuticals should be differently combined in a personalized strategy to tackle such a public health burden. Altered brown adipose tissue (BAT) function contributes to the pathophysiology of obesity and glucose metabolism dysfunctions. BAT thermogenic activity burns glucose and fatty acids to produce heat through uncoupled respiration, and can dissipate the excessive calorie intake, reduce glycemia and circulate fatty acids released from white adipose tissue. Thus, BAT activity is expected to contribute to whole body energy homeostasis and protect against obesity, diabetes and alterations in lipid profile. To date, pharmacological therapies aimed at activating brown fat have failed in clinical trials, due to cardiovascular side effects or scarce efficacy. On the other hand, several studies have identified plant-derived chemical compounds capable of stimulating BAT thermogenesis in animal models, suggesting the translational applications of dietary supplements to fight adipose tissue dysfunctions. This review describes several nutraceuticals with thermogenic properties and provides indications, at a molecular level, of the regulation of the adipocyte thermogenesis by the mentioned phytochemicals.
Collapse
Affiliation(s)
- Andrea Armani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
- Correspondence:
| | - Alessandra Feraco
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Elisabetta Camajani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Stefania Gorini
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Mauro Lombardo
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Massimiliano Caprio
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| |
Collapse
|
16
|
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
|
17
|
Martinez-Sanchez N, Sweeney O, Sidarta-Oliveira D, Caron A, Stanley SA, Domingos AI. The sympathetic nervous system in the 21st century: Neuroimmune interactions in metabolic homeostasis and obesity. Neuron 2022; 110:3597-3626. [PMID: 36327900 PMCID: PMC9986959 DOI: 10.1016/j.neuron.2022.10.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/23/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
The sympathetic nervous system maintains metabolic homeostasis by orchestrating the activity of organs such as the pancreas, liver, and white and brown adipose tissues. From the first renderings by Thomas Willis to contemporary techniques for visualization, tracing, and functional probing of axonal arborizations within organs, our understanding of the sympathetic nervous system has started to grow beyond classical models. In the present review, we outline the evolution of these findings and provide updated neuroanatomical maps of sympathetic innervation. We offer an autonomic framework for the neuroendocrine loop of leptin action, and we discuss the role of immune cells in regulating sympathetic terminals and metabolism. We highlight potential anti-obesity therapeutic approaches that emerge from the modern appreciation of SNS as a neural network vis a vis the historical fear of sympathomimetic pharmacology, while shifting focus from post- to pre-synaptic targeting. Finally, we critically appraise the field and where it needs to go.
Collapse
Affiliation(s)
| | - Owen Sweeney
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Davi Sidarta-Oliveira
- Physician-Scientist Graduate Program, Obesity and Comorbidities Research Center, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ana I Domingos
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.
| |
Collapse
|
18
|
Pileggi C, Hooks B, McPherson R, Dent R, Harper ME. Targeting skeletal muscle mitochondrial health in obesity. Clin Sci (Lond) 2022; 136:1081-1110. [PMID: 35892309 PMCID: PMC9334731 DOI: 10.1042/cs20210506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/26/2022] [Accepted: 07/05/2022] [Indexed: 11/21/2022]
Abstract
Metabolic demands of skeletal muscle are substantial and are characterized normally as highly flexible and with a large dynamic range. Skeletal muscle composition (e.g., fiber type and mitochondrial content) and metabolism (e.g., capacity to switch between fatty acid and glucose substrates) are altered in obesity, with some changes proceeding and some following the development of the disease. Nonetheless, there are marked interindividual differences in skeletal muscle composition and metabolism in obesity, some of which have been associated with obesity risk and weight loss capacity. In this review, we discuss related molecular mechanisms and how current and novel treatment strategies may enhance weight loss capacity, particularly in diet-resistant obesity.
Collapse
Affiliation(s)
- Chantal A. Pileggi
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, Canada, K1H 8M5
- Ottawa Institute of Systems Biology, University of Ottawa, ON, Canada, K1H 8M5
| | - Breana G. Hooks
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, Canada, K1H 8M5
- Ottawa Institute of Systems Biology, University of Ottawa, ON, Canada, K1H 8M5
| | - Ruth McPherson
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Robert R.M. Dent
- Division of Endocrinology, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, Canada, K1H 8M5
- Ottawa Institute of Systems Biology, University of Ottawa, ON, Canada, K1H 8M5
| |
Collapse
|
19
|
Zhao Z, Yang R, Li M, Bao M, Huo D, Cao J, Speakman JR. Effects of ambient temperatures between 5 and 35 oC on energy balance, body mass and body composition in mice. Mol Metab 2022; 64:101551. [PMID: 35870706 PMCID: PMC9382332 DOI: 10.1016/j.molmet.2022.101551] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022] Open
Abstract
Background Considerable attention is currently focused on the potential to switch on brown adipose tissue (BAT), or promote browning of white adipose tissue, to elevate energy expenditure and thereby reduce obesity levels. These processes are already known to be switched on by cold exposure. Yet humans living in colder regions do not show lower levels of obesity. This could be because humans shield themselves from external temperatures, or because the resultant changes in BAT and thermogenesis are offset by elevated food intake, or reductions in other components of expenditure. Scope of Review We exposed mice to 11 different ambient temperatures between 5 and 35 °C and characterized their energy balance and body weight/composition. As it got colder mice progressively increased their energy expenditure coincident with changes in thyroid hormone levels and increased BAT activity. Simultaneously, these increases in expenditure were matched by elevated food intake, and body mass remained stable. Nevertheless, within this envelope of unchanged body mass there were significant changes in body composition – with increases in the sizes of the liver and small intestine, presumably to support the greater food intake, and reductions in the level of stored fat – maximally providing about 10% of the total elevated energy demands. Major Conclusions Elevating activity of BAT may be a valid strategy to reduce fat storage even if overall body mass is unchanged but if it is mostly offset by elevated food intake, as found here, then the impacts may be small. Male and female mice were exposed to 11 different ambient temperatures between 5 and 35 °C. As it got colder mice increased both energy expenditure and food intake. Increased energy expenditure was coincident with increased THs and BAT activity. Stored fat was considerably reduced in colder conditions, providing about 10% of the elevated energy requirements. Elevating activity of BAT may be a valid strategy to reduce fat storage.
Collapse
Affiliation(s)
- Zhijun Zhao
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China.
| | - Rui Yang
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - Min Li
- Shenzhen key laboratory of metabolic health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Menghuan Bao
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - Daliang Huo
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - Jing Cao
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - John R Speakman
- Shenzhen key laboratory of metabolic health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100100, China.
| |
Collapse
|
20
|
Monfort-Pires M, Regeni-Silva G, Dadson P, Nogueira GA, U-Din M, Ferreira SRG, Sapienza MT, Virtanen K, Velloso LA. Brown fat triglyceride content is associated with cardiovascular risk markers in adults from a tropical region. Front Endocrinol (Lausanne) 2022; 13:919588. [PMID: 35928901 PMCID: PMC9343995 DOI: 10.3389/fendo.2022.919588] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Brown adipose tissue (BAT) is regarded as an interesting potential target for the treatment of obesity, diabetes, and cardiovascular diseases, and the detailed characterization of its structural and functional phenotype could enable an advance in these fields. Most studies evaluating BAT structure and function were performed in temperate climate regions, and we are yet to know how these findings apply to the 40% of the world's population living in tropical areas. Here, we used 18F-fluorodeoxyglucose positron emission tomography - magnetic resonance imaging to evaluate BAT in 45 lean, overweight, and obese volunteers living in a tropical area in Southeast Brazil. We aimed at investigating the associations between BAT activity, volume, metabolic activity, and BAT content of triglycerides with adiposity and cardiovascular risk markers in a sample of adults living in a tropical area and we showed that BAT glucose uptake is not correlated with leanness; instead, BAT triglyceride content is correlated with visceral adiposity and markers of cardiovascular risk. This study expands knowledge regarding the structure and function of BAT in people living in tropical areas. In addition, we provide evidence that BAT triglyceride content could be an interesting marker of cardiovascular risk.
Collapse
Affiliation(s)
- Milena Monfort-Pires
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University ofCampinas (UNICAMP), Campinas, Brazil
| | - Giulianna Regeni-Silva
- Department of Nutrition, School of Public Health -University of São Paulo, São Paulo, Brazil
| | - Prince Dadson
- Turku PET Centre, University of Turku, Turku, Finland
| | - Guilherme A. Nogueira
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University ofCampinas (UNICAMP), Campinas, Brazil
| | - Mueez U-Din
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Sandra R. G. Ferreira
- Department of Epidemiology, School of Public Health-University of São Paulo, São Paulo, Brazil
| | - Marcelo Tatit Sapienza
- Division of Nuclear Medicine, Department of Radiology and Oncology, Medical School of University of São Paulo (FMUSP), São Paulo, Finland
| | - Kirsi A. Virtanen
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland (UEF), Kuopio, Finland
| | - Licio A. Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University ofCampinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
21
|
Chi ZC. Metabolic associated fatty liver disease is a disease related to sympathetic nervous system activation. Shijie Huaren Xiaohua Zazhi 2022; 30:465-476. [DOI: 10.11569/wcjd.v30.i11.465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Strong evidence from animal and human studies shows that sympathetic nervous system (SNS) activation is a key factor in the development of metabolic associated fatty liver disease (MAFLD). Activation of the sympathetic nervous system plays an important role in the pathogenesis of obesity, metabolic syndrome, diabetes, hypertension, and MAFLD. When genetically susceptible subjects are exposed to a variety of epigenetic changes, their liver damage may develop into MAFLD. Thus, the pathogenesis of MAFLD is complex, involving the complex interaction of insulin resistance, abnormal hormone secretion, obesity, diet, genetic factors, immune activation, gut microbiota, and other factors. In these processes, the role of sympathetic nerves cannot be underestimated. Notably, SNS has been proposed as a therapeutic target for MAFLD by inhibiting sympathetic nerves. It is worthy of further discussion and research.
Collapse
Affiliation(s)
- Zhao-Chun Chi
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao 266011, Shandong Province, China
| |
Collapse
|
22
|
Extract of Ephedra sinica Stapf Induces Browning of Mouse and Human White Adipocytes. Foods 2022; 11:foods11071028. [PMID: 35407115 PMCID: PMC8998140 DOI: 10.3390/foods11071028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 11/23/2022] Open
Abstract
Browning of adipocytes using herbal extracts is an attractive and realistic strategy for obesity treatment. Ephedra sinica Stapf (E. sinica) is an Asian traditional medicine known to activate brown adipocytes. To evaluate the effect of E. sinica (EEs) on the browning of white adipocytes, expression levels of browning markers, including uncoupling protein 1 (UCP1), were determined using qPCR, Western blot, and immunocytochemistry after mature mouse inguinal preadipocyte (mIPA) and human adipose-derived stem cells (hADSCs) were treated with EEs. In addition, mitochondrial activity was determined by analyzing MitoTracker staining, mtDNA copy number, and oxygen consumption rate (OCR). Treatment with EEs suppressed lipid accumulation and expression levels of adipogenic markers, including Pparg, during mIPA differentiation. In mature mIPA and hADSCs browning markers, including Ucp1, were up-regulated by EEs. In addition, EEs increased expression of mitochondrial genes, mtDNA copy number, and OCR. EEs showed a dual function: inhibiting adipogenesis in immature preadipocytes, and promoting thermogenesis via browning in mature white adipocytes. Therefore, E. sinica is a potential herb for regulating energy metabolism by inducing the browning process.
Collapse
|
23
|
McKie GL, Medak KD, Shamshoum H, Wright DC. Topical application of the pharmacological cold mimetic menthol stimulates brown adipose tissue thermogenesis through a TRPM8, UCP1, and norepinephrine dependent mechanism in mice housed at thermoneutrality. FASEB J 2022; 36:e22205. [PMID: 35157333 DOI: 10.1096/fj.202101905rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/29/2022]
Abstract
Increasing whole-body energy expenditure via the pharmacological activation of uncoupling protein 1 (UCP1)-dependent brown adipose tissue (BAT) thermogenesis is a promising weight management strategy, yet most therapeutics studied in rodents to date either induce compensatory increases in energy intake, have thermogenic effects that are confounded by sub-thermoneutral housing temperatures or are not well tolerated in humans. Here, we sought to determine whether the non-invasive topical application of the pharmacological cold mimetic and transient receptor potential (TRP) cation channel subfamily M member 8 (TRPM8) agonist L-menthol (MNTH), could be used to stimulate BAT thermogenesis and attenuate weight gain in mice housed at thermoneutrality. Using three different strains of mice and multiple complimentary approaches to quantify thermogenesis in vivo, coupled with ex vivo models to quantify direct thermogenic effects, we were able to convincingly demonstrate the following: (1) acute topical MNTH application induces BAT thermogenesis in a TRPM8- and UCP1-dependent manner; (2) MNTH-induced BAT thermogenesis is sufficient to attenuate weight gain over time without affecting energy intake in lean and obese mice; (3) the ability of topical MNTH application to stimulate BAT thermogenesis is mediated, in part, by a central mechanism involving the release of norepinephrine. These data collectively suggest that topical application of MNTH may be a promising weight management strategy.
Collapse
Affiliation(s)
- Greg L McKie
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Kyle D Medak
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Hesham Shamshoum
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - David C Wright
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
24
|
Kulterer OC, Herz CT, Prager M, Schmöltzer C, Langer FB, Prager G, Marculescu R, Kautzky-Willer A, Hacker M, Haug AR, Kiefer FW. Brown Adipose Tissue Prevalence Is Lower in Obesity but Its Metabolic Activity Is Intact. Front Endocrinol (Lausanne) 2022; 13:858417. [PMID: 35432192 PMCID: PMC9009254 DOI: 10.3389/fendo.2022.858417] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/21/2022] [Indexed: 01/09/2023] Open
Abstract
Due to its high metabolic activity, brown adipose tissue (BAT) has become a promising target for the development of novel treatment concepts for metabolic disease. Despite several reports of a negative association between the presence of active BAT and obesity, very little is known about the quantitative and qualitative differences of BAT in lean and obese individuals. Systematic studies directly comparing cold-induced BAT activity in leanness and obesity are currently lacking. Here we studied BAT mass and function in 31 lean and 64 obese men and women. After a standardized cooling protocol using a water-perfused vest, 18F-FDG-positron emission tomography/computed tomography scans were performed, and BAT was delineated using lean body-mass adjusted standardized uptake value (SUV) thresholds in anatomic regions with fat radiodensity. Cold-induced thermogenesis (CIT), a functional readout of BAT activity, was quantified by indirect calorimetry. Active BAT was present in a significantly higher proportion of lean than obese individuals (58% vs. 33%, p=0.019). In these participants with active BAT, however, BAT volume and activity did not differ between leanness and obesity. Accordingly, CIT was similar in both weight groups. BAT metrics were not related to adiposity or total fat mass per se. However, in obese participants a strong negative correlation existed between visceral adipose tissue and BAT volume, 18F-FDG uptake and CIT. In summary, despite a significantly lower prevalence of BAT, the metabolic activity and thermogenic capacity of BAT appears to be still intact in obesity and is inversely associated with visceral fat mass.
Collapse
Affiliation(s)
- Oana C. Kulterer
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Carsten T. Herz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Marlene Prager
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Christoph Schmöltzer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Felix B. Langer
- Division of Visceral Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Gerhard Prager
- Division of Visceral Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Rodrig Marculescu
- Division of Medical-Chemical Laboratory Diagnostics, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Alexandra Kautzky-Willer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Alexander R. Haug
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Florian W. Kiefer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- *Correspondence: Florian W. Kiefer,
| |
Collapse
|
25
|
Chen KY, Brychta RJ, Israni NS, Jiang A, Lea HJ, Lentz TN, Pierce AE, Cypess AM. Activating Human Adipose Tissue with the β3-Adrenergic Agonist Mirabegron. Methods Mol Biol 2022; 2448:83-96. [PMID: 35167091 DOI: 10.1007/978-1-0716-2087-8_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
An appealing strategy for treatment of metabolic disease in humans is activation of brown adipose tissue (BAT), a thermogenic organ best visualized through 18F-FDG PET/CT. BAT has been activated to varying degrees by mild cold exposure. However, this approach can cause undesirable stress, and there remains no consensus protocol. Here, we describe standardized methods for both acute and chronic activation of BAT using the orally administered β3-adrenergic receptor (AR) agonist, mirabegron. Acute pharmacological stimulation has enabled quantification of whole-body BAT volume and metabolic activity using PET/CT imaging, and chronic stimulation increases these properties of BAT over time.
Collapse
Affiliation(s)
- Kong Y Chen
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA.
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Robert J Brychta
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nikita S Israni
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alex Jiang
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hannah J Lea
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Taylor N Lentz
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anne E Pierce
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes, Bethesda, MD, USA.
- Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
26
|
Almeida DL, Moreira VM, Cardoso LE, Junior MDF, Pavanelo A, Ribeiro TA, da Silva Franco CC, Tófolo LP, Peres MNC, Ribeiro MVG, Ferreira ARO, Gomes RM, Miranda RA, Trevenzoli IH, Armitage JA, Palma-Rigo K, de Freitas Mathias PC. Lean in one way, in obesity another: effects of moderate exercise in brown adipose tissue of early overfed male Wistar rats. Int J Obes (Lond) 2022; 46:137-143. [PMID: 34552207 DOI: 10.1038/s41366-021-00969-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Early postnatal overfeeding (PO) induces long-term overweight and reduces brown adipose tissue (BAT) thermogenesis. Exercise has been suggested as a possible intervention to increase BAT function. In this study, we investigated chronical effects of moderate-intensity exercise in BAT function in postnatal overfed male Wistar rats METHODS: Litters' delivery was on postnatal-day 0 - PN0. At PN2, litters were adjusted to nine (normal litter - NL) or three pups (small litter - SL) per dam. Animals were weaned on PN21 and in PN30 randomly divided into sedentary (NL-Sed and SL-Sed) or exercised (NL-Exe and SL-Exe), N of 14 litters per group. Exercise protocol started (PN30) with an effort test; training sessions were performed three times weekly at 60% of the VO2max achieved in effort test, until PN80. On PN81, a temperature transponder was implanted beneath the interscapular BAT, whose temperature was assessed in periods of lights-on and -off from PN87 to PN90. Sympathetic nerve activation of BAT was registered at PN90. Animals were euthanized at PN91 and tissues collected RESULTS: PO impaired BAT thermogenesis in lights-on (pPO < 0.0001) and -off (pPO < 0.01). Exercise increased BAT temperature in lights-on (pExe < 0.0001). In NL-Exe, increased BAT activity was associated with higher sympathetic activity (pExe < 0.05), β3-AR (pExe < 0.001), and UCP1 (pExe < 0.001) content. In SL-Exe, increasing BAT thermogenesis is driven by a combination of tissue morphology remodeling (pExe < 0.0001) with greater effect in increasing UCP1 (pExe < 0.001) and increased β3-AR (pExe < 0.001) content. CONCLUSION Moderate exercise chronically increased BAT thermogenesis in both, NL and SL groups. In NL-Exe by increasing Sympathetic activity, and in SL-Exe by a combination of increased β3-AR and UCP1 content with morphologic remodeling of BAT. Chronically increasing BAT thermogenesis in obese subjects may lead to higher overall energy expenditure, favoring the reduction of obesity and related comorbidities.
Collapse
Affiliation(s)
- Douglas Lopes Almeida
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil.
- Department of Physiology, State University of Londrina, Londrina, Paraná, Brazil.
| | - Veridiana Mota Moreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
- Department of Physiology, State University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Lucas Eduardo Cardoso
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | | | - Audrei Pavanelo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Claudinéia Conationi da Silva Franco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Laize Perón Tófolo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Maria Natália Chimirri Peres
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Anna Rebeka Oliveira Ferreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Rodrigo Mello Gomes
- Physiological Sciences Department, Federal University of Goiás, Av Esperança, Goiânia/GO, Brazil
| | - Rosiane Aparecida Miranda
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, 550 Av, Pedro Calmon, Rio de Janeiro, Brazil
| | - Isis Hara Trevenzoli
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, 550 Av, Pedro Calmon, Rio de Janeiro, Brazil
| | - James Andrew Armitage
- Deakin University, School of Medicine, Optometry, 75 Pigdons Rd, Waurn Ponds, VIC, Australia
| | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Paulo Cesar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| |
Collapse
|
27
|
Liu X, Zhang Z, Song Y, Xie H, Dong M. An update on brown adipose tissue and obesity intervention: Function, regulation and therapeutic implications. Front Endocrinol (Lausanne) 2022; 13:1065263. [PMID: 36714578 PMCID: PMC9874101 DOI: 10.3389/fendo.2022.1065263] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
Overweight and obesity have become a world-wide problem. However, effective intervention approaches are limited. Brown adipose tissue, which helps maintain body temperature and contributes to thermogenesis, is dependent on uncoupling protein1. Over the last decade, an in-creasing number of studies have found that activating brown adipose tissue and browning of white adipose tissue can protect against obesity and obesity-related metabolic disease. Brown adipose tissue has gradually become an appealing therapeutic target for the prevention and re-versal of obesity. However, some important issues remain unresolved. It is not certain whether increasing brown adipose tissue activity is the cause or effect of body weight loss or what the risks might be for sympathetic nervous system-dependent non-shivering thermogenesis. In this review, we comprehensively summarize approaches to activating brown adipose tissue and/or browning white adipose tissue, such as cold exposure, exercise, and small-molecule treatment. We highlight the functional mechanisms of small-molecule treatment and brown adipose tissue transplantation using batokine, sympathetic nervous system and/or gut microbiome. Finally, we discuss the causality between body weight loss induced by bariatric surgery, exercise, and brown adipose tissue activity.
Collapse
Affiliation(s)
- Xiaomeng Liu
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
- Department of Nutrition and Food Hygiene, College of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhi Zhang
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yajie Song
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Hengchang Xie
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- *Correspondence: Meng Dong, ; Hengchang Xie,
| | - Meng Dong
- Department of Nutrition and Food Hygiene, College of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- *Correspondence: Meng Dong, ; Hengchang Xie,
| |
Collapse
|
28
|
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
|
29
|
Gad MZ, Azab SS, Khattab AR, Farag MA. Over a century since ephedrine discovery: an updated revisit to its pharmacological aspects, functionality and toxicity in comparison to its herbal extracts. Food Funct 2021; 12:9563-9582. [PMID: 34533553 DOI: 10.1039/d1fo02093e] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Ephedrine, a sympathomimetic amine that exhibits several adrenaline actions, is a plant alkaloid that is a common ingredient in several cold, asthma and narcolepsy treatment preparations, and in obesity management and sport medicine. Its principal action mechanism relies on its direct adrenergic actions as well as indirect role that involves the release of epinephrine and norepinephrine, thus increasing the activity of epinephrine and norepinephrine at the postsynaptic α and β receptors. Nevertheless, its serious side effects, including stroke, heart attack, drug abuse and interactions, have never been comprehensively reviewed. We conducted a systematic review of data on ephedrine, including its occurrence in functional foods, pharmacological aspects, metabolism, pharmaco/toxicokinetics and clinical features. Furthermore, a review of ephedrine natural structural analogues with regards to their differential adrenergic receptor binding affinities, food interaction, and their impact on the pharmacokinetics and effects relative to ephedrine are presented for the first time, and in comparison to its action when present in herbs.
Collapse
Affiliation(s)
- Mohamed Z Gad
- Department of Biochemistry, Faculty of Pharmacy & Biotechnology, The German University in Cairo, Egypt
| | - Samar S Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amira R Khattab
- Pharmacognosy Department, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria 1029, Egypt
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr El Aini St, P.B. 11562, Cairo, Egypt. .,Chemistry Department, School of Sciences & Engineering, The American University in Cairo, New Cairo 11835, Egypt
| |
Collapse
|
30
|
Abstract
The hypothalamic–pituitary–adrenal axis is a tightly regulated system that represents one of the body’s mechanisms for responding to acute and chronic stress. Prolonged stress and/or inadequate regulation of the stress system can lead to a condition of chronic hypercortisolism or, in some cases, a blunted cortisol response to stress, contributing to insulin resistance, increased adiposity and type 2 diabetes mellitus. Moreover, acute and chronic stress can exacerbate or worsen metabolic conditions by supporting an inflammatory state and a tight relationship between stress, inflammation and adipose tissue has been reported and has been a growing subject of interest in recent years. We reviewed and summarized the evidence supporting hypothalamic–pituitary–adrenal axis dysregulation as an important biological link between stress, obesity, inflammation and type 2 diabetes mellitus. Furthermore, we emphasized the possible role of infectious-related stress such as SarsCov2 infection in adrenal axis dysregulation, insulin resistance and diabetes in a bidirectional link. Understanding and better defining the links between stress and obesity or diabetes could contribute to further definition of the pathogenesis and the management of stress-related complications, in which the HPA axis dysregulation has a primary role.
Collapse
|
31
|
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
|
32
|
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
|
33
|
Yan H, Qian G, Yang R, Luo Z, Wang X, Xie T, Zhao X, Shan J. Huanglong Antitussive Granule Relieves Acute Asthma Through Regulating Pulmonary Lipid Homeostasis. Front Pharmacol 2021; 12:656756. [PMID: 33967801 PMCID: PMC8103164 DOI: 10.3389/fphar.2021.656756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/25/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Asthma is a respiratory disease with chronic airway inflammatory, and individuals with asthma exacerbations is one of the most frequent causes of hospitalization. Huanglong antitussive granule (HL Granule), a Chinese proprietary herbal medicine, has been proved to be effective in the clinical treatment of pulmonary disease. This study is devoted to the pharmacodynamics of HL Granule in acute asthma and the possible mechanism from the perspective of lipidomics. Methods: Mice were divided into four groups, control group, acute asthma model group, HL Granule treatment and montelukast sodium treatment group. Acute asthma was induced by ovalbumin (OVA). Histopathology, pulmonary function and enzyme linked immunosorbent assay (ELISA) were used to validated model and effect of HL Granule. Lipids were detected by ultra-high-performance liquid chromatography coupled to hybrid Quadrupole-Exactive Orbitrap mass spectrometry (UHPLC-Q-Exactive Orbitrap MS) and identified by MS-DAIL and built-in Lipidblast database. Differentially expressed lipids recalled in HL Granule treatment group were extracted for heatmap, enrichment analysis and correlation analysis. Results: HL Granule was effective in decreasing airway hyperresponsiveness (AHR), airway inflammatory and the levels of IL-4 and IL-5. A total of 304 and 167 lipids were identified in positive and negative ion mode, respectively. Among these, 104 and 73 lipids were reserved in HL Granule group (FDR < 0.05), including acylcarnitine (ACar), fatty acid (FA), lysophosphatidylcholine (LPC), phosphatidylcholine (PC), lysophosphatidylethanolamine (LPE), phosphatidylethanolamine (PE), phosphatidylglycerol (PG), phosphatidylinositol (PI), phosphatidylserine (PS), diglyceride (DG), triglyceride (TG), sphingomyelin (SM) and ceramide (Cer). Furthermore, 118 and 273 correlations among 47 and 96 lipids in the positive and negative were observed, with ether-linked phosphatidylethanolamine (PEe) and phosphatidylcholine (PCe) (FDR < 0.001, Spearman correlation coefficient r 2 > 0.75). Conclusion: HL Granule might improve pulmonary lipid homeostasis and could be used as an alternative or supplementary therapy in clinical for the treatment of asthma.
Collapse
Affiliation(s)
- Hua Yan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Guiying Qian
- Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Rui Yang
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zichen Luo
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xianzheng Wang
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Tong Xie
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xia Zhao
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
34
|
Van Schaik L, Kettle C, Green R, Irving HR, Rathner JA. Effects of Caffeine on Brown Adipose Tissue Thermogenesis and Metabolic Homeostasis: A Review. Front Neurosci 2021; 15:621356. [PMID: 33613184 PMCID: PMC7889509 DOI: 10.3389/fnins.2021.621356] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
The impact of brown adipose tissue (BAT) metabolism on understanding energy balance in humans is a relatively new and exciting field of research. The pathogenesis of obesity can be largely explained by an imbalance between caloric intake and energy expenditure, but the underlying mechanisms are far more complex. Traditional non-selective sympathetic activators have been used to artificially elevate energy utilization, or suppress appetite, however undesirable side effects are apparent with the use of these pharmacological interventions. Understanding the role of BAT, in relation to human energy homeostasis has the potential to dramatically offset the energy imbalance associated with obesity. This review discusses paradoxical effects of caffeine on peripheral adenosine receptors and the possible role of adenosine in increasing metabolism is highlighted, with consideration to the potential of central rather than peripheral mechanisms for caffeine mediated BAT thermogenesis and energy expenditure. Research on the complex physiology of adipose tissue, the embryonic lineage and function of the different types of adipocytes is summarized. In addition, the effect of BAT on overall human metabolism and the extent of the associated increase in energy expenditure are discussed. The controversy surrounding the primary β-adrenoceptor involved in human BAT activation is examined, and suggestions as to the lack of translational findings from animal to human physiology and human in vitro to in vivo models are provided. This review compares and distinguishes human and rodent BAT effects, thus developing an understanding of human BAT thermogenesis to aid lifestyle interventions targeting obesity and metabolic syndrome. The focus of this review is on the effect of BAT thermogenesis on overall metabolism, and the potential therapeutic effects of caffeine in increasing metabolism via its effects on BAT.
Collapse
Affiliation(s)
- Lachlan Van Schaik
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
| | - Christine Kettle
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
| | - Rodney Green
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
| | - Helen R. Irving
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
| | - Joseph A. Rathner
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
- Department of Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
35
|
Functional characterization of human brown adipose tissue metabolism. Biochem J 2020; 477:1261-1286. [PMID: 32271883 DOI: 10.1042/bcj20190464] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 02/07/2023]
Abstract
Brown adipose tissue (BAT) has long been described according to its histological features as a multilocular, lipid-containing tissue, light brown in color, that is also responsive to the cold and found especially in hibernating mammals and human infants. Its presence in both hibernators and human infants, combined with its function as a heat-generating organ, raised many questions about its role in humans. Early characterizations of the tissue in humans focused on its progressive atrophy with age and its apparent importance for cold-exposed workers. However, the use of positron emission tomography (PET) with the glucose tracer [18F]fluorodeoxyglucose ([18F]FDG) made it possible to begin characterizing the possible function of BAT in adult humans, and whether it could play a role in the prevention or treatment of obesity and type 2 diabetes (T2D). This review focuses on the in vivo functional characterization of human BAT, the methodological approaches applied to examine these features and addresses critical gaps that remain in moving the field forward. Specifically, we describe the anatomical and biomolecular features of human BAT, the modalities and applications of non-invasive tools such as PET and magnetic resonance imaging coupled with spectroscopy (MRI/MRS) to study BAT morphology and function in vivo, and finally describe the functional characteristics of human BAT that have only been possible through the development and application of such tools.
Collapse
|
36
|
Blondin DP, Nielsen S, Kuipers EN, Severinsen MC, Jensen VH, Miard S, Jespersen NZ, Kooijman S, Boon MR, Fortin M, Phoenix S, Frisch F, Guérin B, Turcotte ÉE, Haman F, Richard D, Picard F, Rensen PCN, Scheele C, Carpentier AC. Human Brown Adipocyte Thermogenesis Is Driven by β2-AR Stimulation. Cell Metab 2020; 32:287-300.e7. [PMID: 32755608 DOI: 10.1016/j.cmet.2020.07.005] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 04/10/2020] [Accepted: 07/13/2020] [Indexed: 10/23/2022]
Abstract
Stimulation of brown adipose tissue (BAT) thermogenesis in humans has emerged as an attractive target to improve metabolic health. Pharmacological stimulations targeting the β3-adrenergic receptor (β3-AR), the adrenergic receptor believed to mediate BAT thermogenesis, have historically performed poorly in human clinical trials. Here we report that, in contrast to rodents, human BAT thermogenesis is not mediated by the stimulation of β3-AR. Oral administration of the β3-AR agonist mirabegron only elicited increases in BAT thermogenesis when ingested at the maximal allowable dose. This led to off-target binding to β1-AR and β2-AR, thereby increasing cardiovascular responses and white adipose tissue lipolysis, respectively. ADRB2 was co-expressed with UCP1 in human brown adipocytes. Pharmacological stimulation and inhibition of the β2-AR as well as knockdown of ADRB1, ADRB2, or ADRB3 in human brown adipocytes all confirmed that BAT lipolysis and thermogenesis occur through β2-AR signaling in humans (ClinicalTrials.govNCT02811289).
Collapse
Affiliation(s)
- Denis P Blondin
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Department of Physiology-Pharmacology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Soren Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Righospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Eline N Kuipers
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Mai C Severinsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Righospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Verena H Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Righospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Stéphanie Miard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
| | - Naja Z Jespersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Righospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Mariëtte R Boon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Mélanie Fortin
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Serge Phoenix
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Department of Nuclear Medicine and Radiobiology, Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Frédérique Frisch
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Brigitte Guérin
- Department of Nuclear Medicine and Radiobiology, Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Éric E Turcotte
- Department of Nuclear Medicine and Radiobiology, Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - François Haman
- Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Denis Richard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
| | - Frédéric Picard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Righospitalet, University Hospital of Copenhagen, Copenhagen, Denmark.
| | - André C Carpentier
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
37
|
Schnabl K, Li Y, Klingenspor M. The gut hormone secretin triggers a gut-brown fat-brain axis in the control of food intake. Exp Physiol 2020; 105:1206-1213. [PMID: 32271980 DOI: 10.1113/ep087878] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/06/2020] [Indexed: 12/25/2022]
Abstract
NEW FINDINGS What is the topic of this review? Brown fat's role in meal-associated thermogenesis and the related consequences for energy balance regulation with a focus on the gut hormone secretin, which has been identified as the endocrine molecular mediator of meal-associated brown fat thermogenesis. What advances does it highlight? The finding of the secretin-induced gut-brown fat-brain axis creates new opportunities to manipulate brown fat and thereby energy balance in a natural way while living in a thermoneutral environment. The role of brown fat as a mere catabolic heater organ needs to be revised and more attention should be directed towards the regulatory role of brown fat beyond energy expenditure. ABSTRACT Brown fat research concentrates on the energy expenditure function of this heating organ, whereas previous evidence for a role of brown fat in regulating energy intake has been mostly neglected. Ingestion of a single mixed meal activates human brown fat thermogenesis to the same degree as cold. In mice, activation of brown fat thermogenesis with a β3 -adrenergic receptor agonist inhibits food intake. Pharmacological β-blockade, however, inhibits neither meal-associated thermogenesis nor food intake. We recently identified the gut hormone secretin as a non-adrenergic activator of brown fat. In vivo, secretin treatment acutely increases energy expenditure and inhibits food intake in wild-type, but not in uncoupling protein 1 (UCP1)-knockout (KO) mice, which lack thermogenic brown fat function. Concurrently, secretin alters gene expression of melanocortinergic peptides of hypothalamic neurons in wild-type mice, but not UCP1-KO. Blocking endogenous secretin with a neutralizing antibody attenuates brown fat thermogenesis during refeeding, increases food intake of mice, and alters ad libitum feeding behaviour. Taken together, these findings demonstrate that secretin triggers an endocrine gut-brown adipose tissue-brain axis in the control of satiation. We hypothesize that meal-associated activation of brown adipose tissue thermogenesis induced by secretin results in a rise in brain temperature and increased melanocortinergic signalling. Taken together, brown fat is not a mere heating organ dissipating excess calories but also involved in gut-brain communication in the control of food intake.
Collapse
Affiliation(s)
- Katharina Schnabl
- TUM School of Life Sciences, Technical University of Munich, Freising, Germany.,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany.,ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Yongguo Li
- TUM School of Life Sciences, Technical University of Munich, Freising, Germany.,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Martin Klingenspor
- TUM School of Life Sciences, Technical University of Munich, Freising, Germany.,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany.,ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| |
Collapse
|
38
|
Liu H, Xu Y, Hu F. AMPK in the Ventromedial Nucleus of the Hypothalamus: A Key Regulator for Thermogenesis. Front Endocrinol (Lausanne) 2020; 11:578830. [PMID: 33071984 PMCID: PMC7538541 DOI: 10.3389/fendo.2020.578830] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/24/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity has become a global health issue, but effective therapies remain very limited. Adaptive thermogenesis promotes weight loss by dissipating energy in the form of heat, thereby representing a promising target to counteract obesity. Notably, the regulation of thermogenesis is tightly orchestrated by complex neuronal networks, especially those in the hypothalamus. Recent evidence highlights the importance of adenosine monophosphate-activated protein kinase (AMPK) within the ventromedial nucleus of the hypothalamus (VMH) in modulating thermogenesis. Various molecules, such as GLP-1, leptin, estradiol, and thyroid hormones, have been reported to act on the VMH to inhibit AMPK, which subsequently increases thermogenesis through the activation of the sympathetic nervous system (SNS). In this review, we summarize the critical role of AMPK within the VMH in the control of energy balance, focusing on its contribution to thermogenesis and the associated mechanisms.
Collapse
Affiliation(s)
- Hailan Liu
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Yong Xu
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Fang Hu
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Fang Hu
| |
Collapse
|
39
|
Miller CT, Fraser SF, Selig SE, Rice T, Grima M, van den Hoek DJ, Ika Sari C, Lambert GW, Dixon JB. Fitness, Strength and Body Composition during Weight Loss in Women with Clinically Severe Obesity: A Randomised Clinical Trial. Obes Facts 2020; 13:307-321. [PMID: 32702706 PMCID: PMC7588734 DOI: 10.1159/000506643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/08/2020] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION To determine whether combined exercise training with an energy-restricted diet leads to improved physical fitness and body composition when compared to energy restriction alone in free-living premenopausal women with clinically severe obesity. METHODS Sixty premenopausal women (BMI of 40.4 ± 6.7) were randomised to energy restriction only (ER) or to exercise plus energy restriction (EXER) for 12 months. Body composition and fitness were measured at baseline, 3, 6 and 12 months. RESULTS VO2 peak improved more for EXER compared to ER at 3 (mean difference ± SEM 2.5 ± 0.9 mL ∙ kg-1 ∙ min-1, p = 0.006) and 6 (3.1 ± 1.2 mL ∙ kg-1 ∙ min-1, p = 0.007) but not 12 months (2.3 ± 1.6 mL ∙ kg-1 ∙ min-1, p = 0.15). Muscle strength improved more for EXER compared to ER at all time points. No differences between groups for lean mass were observed at 12 months. CONCLUSION Combining exercise training with an energy-restricted diet did not lead to greater aerobic power, total body mass, fat mass or limit lean body mass loss at 12 months when compared to energy restriction alone for premenopausal women with clinically severe obesity in free-living situations. Future research should aim to determine an effective lifestyle approach which can be applied in the community setting for this high-risk group.
Collapse
Affiliation(s)
- Clint T Miller
- School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia,
| | - Steve F Fraser
- School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
- Institute for Physical Activity and Nutrition, Deakin University, Geelong, Victoria, Australia
| | - Steve E Selig
- School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Toni Rice
- Clinical Obesity Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Human Neurotransmitters and Clinical Obesity Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Mariee Grima
- Clinical Obesity Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Human Neurotransmitters and Clinical Obesity Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Daniel J van den Hoek
- Institute for Physical Activity and Nutrition, Deakin University, Geelong, Victoria, Australia
- School of Behavioural and Health Sciences, Australian Catholic University, Brisbane, Queensland, Australia
| | - Carolina Ika Sari
- Human Neurotransmitters and Clinical Obesity Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Gavin W Lambert
- Human Neurotransmitters and Clinical Obesity Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Iverson Health Innovation Research Institute and School of Health Sciences, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - John B Dixon
- Clinical Obesity Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Iverson Health Innovation Research Institute and School of Health Sciences, Swinburne University of Technology, Melbourne, Victoria, Australia
| |
Collapse
|
40
|
Qiu Y, Sun L, Hu X, Zhao X, Shi H, Liu Z, Yin X. Compromised browning plasticity of primary subcutaneous adipocytes derived from overweight Chinese adults. Diabetol Metab Syndr 2020; 12:91. [PMID: 33110450 PMCID: PMC7583200 DOI: 10.1186/s13098-020-00599-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022] Open
Abstract
PURPOSE People with obesity have a compromised browning capacity of adipose tissue when faced with sympathetic stimuli. This study aimed to determine whether norepinephrine treatment can enhance the induction of precursor cells from human white adipose tissue to differentiate into adipocytes that express key markers of beige adipocytes, and if there is a difference in this capacity between normal weight and overweight individuals. METHODS Stromal vascular cells derived from subcutaneous white adipose tissue of normal weight and overweight groups were induced to differentiation, with or without norepinephrine, into adipocytes. Oxygen consumption rate, lipolysis, the expression of uncoupling protein 1 and other thermogenic genes were compared between different adiposity and treatment groups. RESULTS Peroxisome proliferator activated receptor γ- coactivator-1 alpha (PGC-1 α) and uncoupling protein 1 gene expression increased significantly in the normal weight group, but not in the overweight group, with norepinephrine treatment. The increments of lipolysis and oxygen consumption rate were also higher in adipocytes from the normal weight group with norepinephrine treatment, as compared with those of the overweight group. PR domain containing protein 16 (PRDM 16) gene expression was higher in the normal weight group compared with that in the overweight group, while there were no significant changes found with norepinephrine treatment in either the normal weight or overweight group. CONCLUSIONS Adipogenic precursor cells derived from overweight individuals were less prone to differentiate into beige-like adipocytes when facing sympathetic stimuli than normal weight ones, resulting in the compromised sympathetic-induced browning capacity in subcutaneous white adipose tissue in overweight individuals, which occurred before the onset of overt obesity.
Collapse
Affiliation(s)
- Yao Qiu
- Department of Surgery, Shandong University Affiliated Jinan Central Hospital, Jinan, Shandong People’s Republic of China
| | - Lizhi Sun
- Department of Clinical Laboratory, Shandong University Affiliated Jinan Central Hospital, Jinan, Shandong People’s Republic of China
| | - Xiaolin Hu
- Department of Endocrinology and Metabolism, Shandong University Affiliated Jinan Central Hospital, Linong road 8, 5-1-801, Jinan, 250013 Shandong People’s Republic of China
| | - Xin Zhao
- Department of Clinical Laboratory, Shandong University Affiliated Jinan Central Hospital, Jinan, Shandong People’s Republic of China
| | - Hongyan Shi
- Department of Surgery, Shandong University Affiliated Jinan Central Hospital, Jinan, Shandong People’s Republic of China
| | - Zhao Liu
- Department of Surgery, Shandong University Affiliated Jinan Central Hospital, Jinan, Shandong People’s Republic of China
| | - Xiao Yin
- Department of Endocrinology and Metabolism, Shandong University Affiliated Jinan Central Hospital, Linong road 8, 5-1-801, Jinan, 250013 Shandong People’s Republic of China
| |
Collapse
|
41
|
Li H, Shen L, Zhang L, Yan B, Sun T, Guo F, Yin X. Reduced Beige Adipogenic Potential in Subcutaneous Adipocytes Derived from Obese Chinese Individuals. Diabetes Metab Syndr Obes 2020; 13:2551-2562. [PMID: 32765034 PMCID: PMC7373414 DOI: 10.2147/dmso.s248112] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 06/26/2020] [Indexed: 01/06/2023] Open
Abstract
PURPOSE Thermogenesis function has made brown/beige adipocyte an attractive target for obesity. Human brown adipose tissue activity is impaired in obesity in vivo. The present study aims to compare the differences in beige adipocyte differentiation potential of subcutaneous adipose tissue derived from normal weight and obese Chinese individuals in vitro. METHODS Adipose-derived stem cells (ADSCs) isolated from subcutaneous fat tissues of normal weight (NW) and obese (OB) groups were induced to differentiate into mature adipocyte with white adipocyte (WA)- and beige adipocyte (BA)-induction treatment. The expression of beige adipocyte marker protein UCP-1 and specific thermogenic genes was detected in differentiated adipocytes via Western blot and rt PCR, and the adipocyte mitochondrial function and lipolysis ability were also measured by oxygen consumption rate (OCR) and glycerol release rate, respectively. RESULTS Either with WA-induction or BA-induction, the expression of UCP-1 and beige adipocyte-specific thermogenic genes in differentiated adipocytes was higher in the NW compared to the OB group, followed by higher OCR and lipolysis ability in NW group than OB group. With BA-induction, expression of UCP-1 and thermogenic genes increased significantly, followed by the increasement in adipocytes OCR and lipolysis rate in NW group compared with WA-induction treatment, but no significant difference was observed in OB group. CONCLUSION Compromised beige adipocyte differentiation plasticity was found in subcutaneous white adipose tissue derived from obese Chinese individuals, which may be due part to the downregulation of β3-adrenergic receptor expression in adipocytes. Discovery of therapeutic agents to active brown adipose tissue through specific pathways could provide a promising approach for treating obesity in the future.
Collapse
Affiliation(s)
- Han Li
- Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong250013, People’s Republic of China
| | - Lin Shen
- Shandong First Medical University, Taian, Shandong271000, People’s Republic of China
| | - Lei Zhang
- Department of Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong250013, People’s Republic of China
| | - Bing Yan
- Department of Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong250013, People’s Republic of China
| | - Tao Sun
- Department of Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong250013, People’s Republic of China
| | - Feng Guo
- Department of Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong250013, People’s Republic of China
| | - Xiao Yin
- Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong250013, People’s Republic of China
- Correspondence: Xiao Yin Email
| |
Collapse
|
42
|
Chen KY, Brychta RJ, Abdul Sater Z, Cassimatis TM, Cero C, Fletcher LA, Israni NS, Johnson JW, Lea HJ, Linderman JD, O'Mara AE, Zhu KY, Cypess AM. Opportunities and challenges in the therapeutic activation of human energy expenditure and thermogenesis to manage obesity. J Biol Chem 2019; 295:1926-1942. [PMID: 31914415 DOI: 10.1074/jbc.rev119.007363] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The current obesity pandemic results from a physiological imbalance in which energy intake chronically exceeds energy expenditure (EE), and prevention and treatment strategies remain generally ineffective. Approaches designed to increase EE have been informed by decades of experiments in rodent models designed to stimulate adaptive thermogenesis, a long-term increase in metabolism, primarily induced by chronic cold exposure. At the cellular level, thermogenesis is achieved through increased rates of futile cycling, which are observed in several systems, most notably the regulated uncoupling of oxidative phosphorylation from ATP generation by uncoupling protein 1, a tissue-specific protein present in mitochondria of brown adipose tissue (BAT). Physiological activation of BAT and other organ thermogenesis occurs through β-adrenergic receptors (AR), and considerable effort over the past 5 decades has been directed toward developing AR agonists capable of safely achieving a net negative energy balance while avoiding unwanted cardiovascular side effects. Recent discoveries of other BAT futile cycles based on creatine and succinate have provided additional targets. Complicating the current and developing pharmacological-, cold-, and exercise-based methods to increase EE is the emerging evidence for strong physiological drives toward restoring lost weight over the long term. Future studies will need to address technical challenges such as how to accurately measure individual tissue thermogenesis in humans; how to safely activate BAT and other organ thermogenesis; and how to sustain a negative energy balance over many years of treatment.
Collapse
Affiliation(s)
- Kong Y Chen
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892.
| | - Robert J Brychta
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Zahraa Abdul Sater
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Thomas M Cassimatis
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Cheryl Cero
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Laura A Fletcher
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Nikita S Israni
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - James W Johnson
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Hannah J Lea
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Joyce D Linderman
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Alana E O'Mara
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Kenneth Y Zhu
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892.
| |
Collapse
|
43
|
Al-Amrani A, AbdelKarim M, AlZabin M, Alzoghaibi M. Low expression of brown and beige fat genes in subcutaneous tissues in obese patients. Arch Med Sci 2019; 15:1113-1122. [PMID: 31572455 PMCID: PMC6764296 DOI: 10.5114/aoms.2018.76684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/20/2018] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION The molecular mechanisms behind obesity pathogenesis remain largely undefined. Impairment in the browning process of subcutaneous tissues proposed to contribute to obesity pathogenesis. In the current study, we aimed to assess whether the expression of brown fat genes in subcutaneous tissues in obese patients is altered as compared to non-obese patients. MATERIAL AND METHODS Participants were recruited from patients undergoing general surgeries. At the same site of surgery, biopsies were taken from the abdominal subcutaneous tissues from each participant, along with a venous blood sample. The expression of BAT genes was measured using a real-time PCR method. Serum FGF21 was measured using an ELISA kit, and the serum blood lipid profile was measured using the Dimension VistaTM 1500 System. RESULTS A total of 58 surgical patients was involved. A low expression of BAT genes was observed in the groups with higher body mass index (BMI) (< 30 kg/m2) as compared to the groups with lower BMI (> 30 kg/m2). The expression of CIDEA and CITED1 was significantly higher in the patients with normal weight as compared to obese (p = 0.01 and p = 0.02, respectively). A significant negative correlation was found between the expression of BAT genes and BMI in patients with BMI < 35 kg/m2. However, the strongest negative correlation was observed in the expression of CIDEA (r = -0.5, p = 0.004), followed by TBX1 (r = -0.4, p = 0.01), CITED1, and ZIC1 (r = -0.4, p = 0.03). Whereas the correlation of UCP1 with BMI remained insignificant (r = -0.29, p = 0.08). When including patients with BMI > 35 kg/m2, the correlation decreased and became insignificant (p = 0.08). No significant correlation was found between the expression of BAT genes and blood lipid profiles (p > 0.05). Serum FGF21 was positively and significantly correlated to the expression of UCP1 (r = 0.56, p = 0.02) and TBX1 (r = 0.62, p = 0.01), however, this correlation was missing in patients with severe obesity. CONCLUSIONS Our data suggested that brown and beige genes expression in abdominal subcutaneous tissues is dysregulated in patients with obesity. Further studies are needed to investigate the role of browning of subcutaneous tissues in regulating body weight and metabolism in human.
Collapse
Affiliation(s)
- Aishah Al-Amrani
- PhD student, Department of Physiology, Faculty of Medicine King Saud University, Riyadh, Saudi Arabia
- Faculty of Applied Medical Sciences, Tabuk University, Tabuk, Saudi Arabia
| | - Mouaadh AbdelKarim
- Department of Physiopathology of Inflammatory Bone Diseases, University of the Littoral, Boulogne sur Mer, France
| | | | - Mohammad Alzoghaibi
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
44
|
Li H, Qi J, Li L. Phytochemicals as potential candidates to combat obesity via adipose non-shivering thermogenesis. Pharmacol Res 2019; 147:104393. [PMID: 31401211 DOI: 10.1016/j.phrs.2019.104393] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 12/23/2022]
Abstract
Obesity is a chronic metabolic disease caused by a long-term imbalance between energy intake and expenditure. The discovery of three different shades of adipose tissues has implications in terms of understanding the pathogenesis and potential interventions for obesity and its related complications. Fat browning, as well as activation of brown adipocytes and new beige adipocytes differentiated from adipogenic progenitor cells, are emerging as interesting and promising methods to curb obesity because of their unique capacity to upregulate non-shivering thermogenesis. This capacity is due to catabolism of stored energy to generate heat through the best characterized thermogenic effector uncoupling protein 1 (UCP1). A variety of phytochemicals have been shown in the literature to contribute to thermogenesis by acting as chemical uncouplers, UCP1 inducers or regulators of fat differentiation and browning. In this review, we summarize the mechanisms and strategies for targeting adipose-mediated thermogenesis and highlight the role of phytochemicals in targeting adipose thermogenesis to fight against obesity. We also discuss proposed targets for how these phytochemical molecules promote BAT activity, WAT browning and beige cell development, thereby offering novel insights into interventional strategies of how phytochemicals may help prevent and manage obesity via adipose thermogenesis.
Collapse
Affiliation(s)
- Hanbing Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, PR China; Section of Endocrinology, School of Medicine, Yale University, New Haven, 06520, USA.
| | - Jiameng Qi
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, PR China
| | - Linghuan Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, PR China
| |
Collapse
|
45
|
Klepac K, Georgiadi A, Tschöp M, Herzig S. The role of brown and beige adipose tissue in glycaemic control. Mol Aspects Med 2019; 68:90-100. [PMID: 31283940 DOI: 10.1016/j.mam.2019.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 12/15/2022]
Abstract
For the past decade, brown adipose tissue (BAT) has been extensively studied as a potential therapy for obesity and metabolic diseases due to its thermogenic and glucose-consuming properties. It is now clear that the function of BAT goes beyond heat production, as it also plays an important endocrine role by secreting the so-called batokines to communicate with other metabolic tissues and regulate systemic energy homeostasis. However, despite numerous studies showing the benefits of BAT in rodents, it is still not clear whether recruitment of BAT can be utilized to treat human patients. Here, we review the advances on understanding the role of BAT in metabolism and its benefits on glucose and lipid homeostasis in both humans and rodents. Moreover, we discuss the latest methodological approaches to assess the contribution of BAT to human metabolism as well as the possibility to target BAT, pharmacologically or by lifestyle adaptations, to treat metabolic disorders.
Collapse
Affiliation(s)
- Katarina Klepac
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Inner Medicine 1, Heidelberg, Germany; Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany
| | - Anastasia Georgiadi
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Inner Medicine 1, Heidelberg, Germany; Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany
| | - Matthias Tschöp
- Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Inner Medicine 1, Heidelberg, Germany; Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany; Chair Molecular Metabolic Control, Technical University Munich, Germany.
| |
Collapse
|
46
|
Lambert GW, Schlaich MP, Eikelis N, Lambert EA. Sympathetic activity in obesity: a brief review of methods and supportive data. Ann N Y Acad Sci 2019; 1454:56-67. [PMID: 31268175 DOI: 10.1111/nyas.14140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/11/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
The increase in the prevalence of obesity and the concomitant rise in obesity-related illness have led to substantial pressure on health care systems throughout the world. While the combination of reduced exercise, increased sedentary time, poor diet, and genetic predisposition is undoubtedly pivotal in generating obesity and increasing disease risk, a large body of work indicates that the sympathetic nervous system (SNS) contributes to obesity-related disease development and progression. In obesity, sympathetic nervous activity is regionalized, with activity in some outflows being particularly sensitive to the obese state, whereas other outflows, or responses to stimuli, may be blunted, thereby making the assessment of sympathetic nervous activation in the clinical setting difficult. Isotope dilution methods and direct nerve recording techniques have been developed and utilized in clinical research, demonstrating that in obesity there is preferential activation of the muscle vasoconstrictor and renal sympathetic outflows. With weight loss, sympathetic activity is reduced. Importantly, sympathetic nervous activity is associated with end-organ dysfunction and changes in sympathetic activation that accompany weight loss are often reflected in an improvement of end-organ function. Whether targeting the SNS directly improves obesity-related illness remains unknown, but merits further attention.
Collapse
Affiliation(s)
- Gavin W Lambert
- The Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, Victoria, Australia.,The School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, University of Western Australia, Perth, Western Australia, Australia
| | - Nina Eikelis
- The Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, Victoria, Australia.,The School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Elisabeth A Lambert
- The Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, Victoria, Australia.,The School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia
| |
Collapse
|
47
|
Liu Y, Fu W, Seese K, Yin A, Yin H. Ectopic brown adipose tissue formation within skeletal muscle after brown adipose progenitor cell transplant augments energy expenditure. FASEB J 2019; 33:8822-8835. [PMID: 31059287 DOI: 10.1096/fj.201802162rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Brown adipose tissue (BAT) thermogenesis increases energy expenditure (EE). Expanding the volume of active BAT via transplantation holds promise as a therapeutic strategy for morbid obesity and diabetes. Brown adipose progenitor cells (BAPCs) can be isolated and expanded to generate autologous brown adipocyte implants. However, the transplantation of brown adipocytes is currently impeded by poor efficiency of BAT tissue formation in vivo and undesirably short engraftment time. In this study, we demonstrated that transplanting BAPCs into limb skeletal muscles consistently led to the ectopic formation of uncoupling protein 1 (UCP1)+pos adipose tissue with long-term engraftment (>4 mo). Combining VEGF with the BAPC transplant further improved BAT formation in muscle. Ectopic engraftment of BAPC-derived BAT in skeletal muscle augmented the EE of recipient mice. Although UCP1 expression declined in long-term BAT grafts, this deterioration can be reversed by swimming exercise because of sympathetic activation. This study suggests that intramuscular transplantation of BAPCs represents a promising approach to deriving functional BAT engraftment, which may be applied to therapeutic BAT transplantation and tissue engineering.-Liu, Y., Fu, W., Seese, K., Yin, A., Yin, H. Ectopic brown adipose tissue formation within skeletal muscle after brown adipose progenitor cell transplant augments energy expenditure.
Collapse
Affiliation(s)
- Yang Liu
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA; and.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Wenyan Fu
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA; and.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Kendall Seese
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Amelia Yin
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA; and.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Hang Yin
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA; and.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
48
|
Evans BA, Merlin J, Bengtsson T, Hutchinson DS. Adrenoceptors in white, brown, and brite adipocytes. Br J Pharmacol 2019; 176:2416-2432. [PMID: 30801689 DOI: 10.1111/bph.14631] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/28/2019] [Accepted: 02/11/2019] [Indexed: 01/01/2023] Open
Abstract
Adrenoceptors play an important role in adipose tissue biology and physiology that includes regulating the synthesis and storage of triglycerides (lipogenesis), the breakdown of stored triglycerides (lipolysis), thermogenesis (heat production), glucose metabolism, and the secretion of adipocyte-derived hormones that can control whole-body energy homeostasis. These processes are regulated by the sympathetic nervous system through actions at different adrenoceptor subtypes expressed in adipose tissue depots. In this review, we have highlighted the role of adrenoceptor subtypes in white, brown, and brite adipocytes in both rodents and humans and have included detailed analysis of adrenoceptor expression in human adipose tissue and clonally derived adipocytes. We discuss important considerations when investigating adrenoceptor function in adipose tissue or adipocytes. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Bronwyn A Evans
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Jon Merlin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, Stockholm, Sweden
| | - Dana S Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| |
Collapse
|
49
|
Osuna-Prieto FJ, Martinez-Tellez B, Sanchez-Delgado G, Aguilera CM, Lozano-Sánchez J, Arráez-Román D, Segura-Carretero A, Ruiz JR. Activation of Human Brown Adipose Tissue by Capsinoids, Catechins, Ephedrine, and Other Dietary Components: A Systematic Review. Adv Nutr 2019; 10:291-302. [PMID: 30624591 PMCID: PMC6416040 DOI: 10.1093/advances/nmy067] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Human brown adipose tissue (BAT) has attracted clinical interest not only because it dissipates energy but also for its potential capacity to counteract obesity and related metabolic disorders (e.g., insulin resistance and dyslipidemia). Cold exposure is the most powerful stimulus for activating and recruiting BAT, and this stimulatory effect is mediated by the transient receptor potential (TRP) channels. BAT can also be activated by other receptors such as the G-protein-coupled bile acid receptor 1 (GPBAR1) or β-adrenergic receptors. Interestingly, these receptors also interact with several dietary components; in particular, capsinoids and tea catechins appear to mimic the effects of cold through a TRP-BAT axis, and they consequently seem to decrease body fat and improve metabolic blood parameters. This systematic review critically addresses the evidence behind the available human studies analyzing the effect of several dietary components (e.g., capsinoids, tea catechins, and ephedrine) on BAT activity. Even though the results of these studies are consistent with the outcomes of preclinical models, the lack of robust study designs makes it impossible to confirm the BAT-activation capacity of the specified dietary components. Further investigation into the effects of dietary components on BAT is warranted to clarify to what extent these components could serve as a powerful strategy to treat obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Francisco J Osuna-Prieto
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, Granada, Spain; Departments of
- Analytical Chemistry, University of Granada, Granada, Spain
- Research and Development of Functional Food Center (CIDAF), Health Sciences Technology Park, Granada, Spain
| | - Borja Martinez-Tellez
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, Granada, Spain; Departments of
- Department of Medicine, Leiden University Medical Center, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden, Netherlands
| | - Guillermo Sanchez-Delgado
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, Granada, Spain; Departments of
| | - Concepción M Aguilera
- Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology, Center for Biomedical Research, University of Granada, Granada, Spain
- CIBEROBN, Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition, Carlos III Health Institute, Madrid, Spain
| | - Jesús Lozano-Sánchez
- Analytical Chemistry, University of Granada, Granada, Spain
- Research and Development of Functional Food Center (CIDAF), Health Sciences Technology Park, Granada, Spain
| | - David Arráez-Román
- Analytical Chemistry, University of Granada, Granada, Spain
- Research and Development of Functional Food Center (CIDAF), Health Sciences Technology Park, Granada, Spain
| | - Antonio Segura-Carretero
- Analytical Chemistry, University of Granada, Granada, Spain
- Research and Development of Functional Food Center (CIDAF), Health Sciences Technology Park, Granada, Spain
| | - Jonatan R Ruiz
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, Granada, Spain; Departments of
| |
Collapse
|
50
|
Loh RKC, Formosa MF, La Gerche A, Reutens AT, Kingwell BA, Carey AL. Acute metabolic and cardiovascular effects of mirabegron in healthy individuals. Diabetes Obes Metab 2019; 21:276-284. [PMID: 30203462 DOI: 10.1111/dom.13516] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/15/2018] [Accepted: 08/24/2018] [Indexed: 12/12/2022]
Abstract
AIMS To quantify acute energy expenditure, supraclavicular skin temperature and cardiovascular responses to four doses of the β3-adrenoceptor agonist, mirabegron. MATERIALS AND METHODS A total of 17 individuals (11 men, six women) participated in this ascending-dose study, receiving single 50-, 100-, 150- and 200-mg doses of mirabegron on four separate days with 3 to 14 days wash-out between each dose. All variables were measured each visit from baseline to 180 minutes post mirabegron treatment. To determine brown adipose tissue (BAT) thermogenic efficacy at each dose, energy expenditure and supraclavicular skin temperature were compared from baseline to 180 minutes post mirabegron treatment. To examine safety, changes in cardiovascular variables at 100, 150 and 200 mg were compared with the standard clinical dose of 50 mg. RESULTS Energy expenditure significantly increased after the 100- (35.6 ± 5.4 kJ/h) and 200-mg (35.6 ± 13.1 kJ/h) doses (P ≤ 0.05), and trended towards an increase after 150 mg (24.1 ± 13.6 kJ/h). Supraclavicular skin temperature increased after 50- (0.22 ± 0.1°C), 100- (0.30 ± 0.1°C) and 150-mg mirabegron doses (0.29 ± 0.1°C; P ≤ 0.05). The change in systolic blood pressure was greater after 150- (7.1 ± 1.3 mm Hg) and 200-mg doses (9.3 ± 1.9 mm Hg) than after the 50-mg dose (2.2 ± 1.3 mm Hg; P ≤ 0.05). The change in heart rate was greater after 200 mg (9.0 ± 2.2 bpm) compared with 50 mg (2.9 ± 1.4 bpm; P ≤ 0.05). CONCLUSIONS A 100-mg dose of mirabegron increases energy expenditure and supraclavicular skin temperature in a β3-adrenoceptor-specific manner, without the off-target elevations in blood pressure or heart rate observed at higher doses.
Collapse
Affiliation(s)
- Rebecca K C Loh
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Melissa F Formosa
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Andre La Gerche
- Sports Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Anne T Reutens
- Clinical Diabetes and Epidemiology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Bronwyn A Kingwell
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Andrew L Carey
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|