1
|
Zhu H, Xu H, Zhang Y, Brodský J, Gablech I, Korabečná M, Neuzil P. Exploring the Frontiers of Cell Temperature Measurement and Thermogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2402135. [PMID: 39467049 DOI: 10.1002/advs.202402135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/24/2024] [Indexed: 10/30/2024]
Abstract
The precise measurement of cell temperature and an in-depth understanding of thermogenic processes are critical in unraveling the complexities of cellular metabolism and its implications for health and disease. This review focuses on the mechanisms of local temperature generation within cells and the array of methods developed for accurate temperature assessment. The contact and noncontact techniques are introduced, including infrared thermography, fluorescence thermometry, and other innovative approaches to localized temperature measurement. The role of thermogenesis in cellular metabolism, highlighting the integral function of temperature regulation in cellular processes, environmental adaptation, and the implications of thermogenic dysregulation in diseases such as metabolic disorders and cancer are further discussed. The challenges and limitations in this field are critically analyzed while technological advancements and future directions are proposed to overcome these barriers. This review aims to provide a consolidated resource for current methodologies, stimulate discussion on the limitations and challenges, and inspire future innovations in the study of cellular thermodynamics.
Collapse
Affiliation(s)
- Hanliang Zhu
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, P. R. China
| | - Haotian Xu
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, P. R. China
| | - Yue Zhang
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, P. R. China
| | - Jan Brodský
- Department of Microelectronics, The Faculty of Electrical Engineering and Communication Technology, Brno University of Technology, Technická 3058/10, Brno, 616 00, Czech Republic
| | - Imrich Gablech
- Department of Microelectronics, The Faculty of Electrical Engineering and Communication Technology, Brno University of Technology, Technická 3058/10, Brno, 616 00, Czech Republic
| | - Marie Korabečná
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Albertov 4, Prague, 128 00, Czech Republic
- Department of Laboratory Medicine, Faculty of Health Care and Social Work, University of Trnava in Trnava, Universitne namestie 1, Trnava, 918 43, Slovakia
| | - Pavel Neuzil
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, P. R. China
| |
Collapse
|
2
|
Musiol E, Fromme T, Hau J, Di Pizio A, Klingenspor M. Comparative functional analysis reveals differential nucleotide sensitivity between human and mouse UCP1. Acta Physiol (Oxf) 2024; 240:e14209. [PMID: 39072954 DOI: 10.1111/apha.14209] [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: 02/28/2023] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
AIM Mitochondrial uncoupling protein 1 (UCP1) is a unique protein of brown adipose tissue. Upon activation by free fatty acids, UCP1 facilitates a thermogenic net proton flux across the mitochondrial inner membrane. Non-complexed purine nucleotides inhibit this fatty acid-induced activity of UCP1. The most available data have been generated from rodent model systems. In light of its role as a putative pharmacological target for treating metabolic disease, in-depth analyses of human UCP1 activity, regulation, and structural features are essential. METHODS In the present study, we established a doxycycline-regulated cell model with inducible human or murine UCP1 expression and conducted functional studies using respirometry comparing wild-type and mutant variants of human UCP1. RESULTS We demonstrate that human and mouse UCP1 exhibit similar specific fatty acid-induced activity but a different inhibitory potential of purine nucleotides. Mutagenesis of non-conserved residues in human UCP1 revealed structural components in α-helix 56 and α-helix 6 crucial for uncoupling function. CONCLUSION Comparative studies of human UCP1 with other orthologs can provide new insights into the structure-function relationship for this mitochondrial carrier and will be instrumental in searching for new activators.
Collapse
Affiliation(s)
- Eva Musiol
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Research Department of Molecular Life Sciences, Technical University of Munich, Freising, Germany
| | - Tobias Fromme
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Research Department of Molecular Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Munich, Germany
| | - Julia Hau
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Research Department of Molecular Life Sciences, Technical University of Munich, Freising, Germany
| | - Antonella Di Pizio
- Molecular Modeling Group, Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Professorship of Chemoinformatics and Protein Modelling, TUM School of Life Sciences, Research Department of Molecular Life Sciences, Technical University of Munich, Freising, Germany
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Research Department of Molecular Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
3
|
Wang S, Lei Y, Wang X, Ma K, Wang C, Sun C, Han T. Association between temperatures and type 2 diabetes: A prospective study in UK Biobank. Diabetes Res Clin Pract 2024; 215:111817. [PMID: 39128563 DOI: 10.1016/j.diabres.2024.111817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
OBJECTIVE This study aims to prospectively examine the association between temperatures and the occurrence of type 2 diabetes (T2D). METHODS We used the CPH models to analyze 103,215 non-diabetic participants in the UK Biobank cohort who answered questions about workplace temperature, to evaluate the survival relationship, and the interaction effects of working environmental temperature and T2D-related genetic risk scores (GRS) on the occurrence of T2D. The occurrence of T2D was assessed by hospital inpatient records. The weighted T2D-related GRS were calculated. RESULTS During 1,355,200.6 person-years follow-up, a total of 2436 participants were documented as having diagnosed T2D. After adjustment, compared to the comfortable group, the participants working in non-comfortable environmental temperature had greater risk of T2D (HR: 1.27, 95 %CI: 1.04 to 1.55, for cold; HR: 1.32, 95 %CI: 1.17 to 1.48 for hot; HR: 1.51, 95 %CI: 1.38 to 1.65 for alternate). Similarly, individuals exposed to different levels of genetic risk scores in alternating hot and cold work environments had a higher risk of developing type 2 diabetes. CONCLUSIONS This study found working in single non-comfortable environmental temperatures was associated with greater risk of T2D occurrence, and exposure to alternating environmental temperatures had the highest risk of range and severity.
Collapse
Affiliation(s)
- ShengYuan Wang
- Department of Occupational Health, School of Public Health, Harbin Medical University, Harbin, PR China
| | - YaTing Lei
- Department of Occupational Health, School of Public Health, Harbin Medical University, Harbin, PR China
| | - XiaoLi Wang
- Department of Occupational Health, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Kun Ma
- Department of Hygiene Toxicology, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Cheng Wang
- Department of Environmental Health, School of Public Health, Harbin Medical University, Harbin, PR China
| | - ChangHao Sun
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150081, PR China.
| | - TianShu Han
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang Province 150081, PR China.
| |
Collapse
|
4
|
Liu W, Zou H, You D, Zhang H, Xu L. Sodium Houttuybonate Promotes the Browning of White Adipose Tissue by Inhibiting Ferroptosis via the AMPK-NRF2-HO1 Pathway. Antioxidants (Basel) 2024; 13:1057. [PMID: 39334717 PMCID: PMC11428211 DOI: 10.3390/antiox13091057] [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: 07/29/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
The rising prevalence of obesity has resulted in an increased demand for innovative and effective treatment strategies. Houttuynia cordata Thunb. (H. cordata) has demonstrated promising potential in preventing obesity. However, the mechanism underlying the anti-obesity effects of H. cordata and its bioactive component, sodium houttuybonate (SH), remains unclear. Our study reveals that SH treatment promotes the browning of inguinal white adipose tissue (iWAT) and prevents the obesity induced by a high-fat diet. SH significantly mitigates ferroptosis by upregulating glutathione peroxidase 4 (Gpx4) and decreasing malondialdehyde (MDA) levels, while also enhancing superoxide dismutase (SOD) levels. Furthermore, SH promotes the phosphorylation of AMP-activated protein kinase (AMPK), which subsequently increases the expression of nuclear factor erythroid 2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1) in the iWAT. However, the effects of SH were attenuated by ML385, an Nrf2 inhibitor. Collectively, our findings suggest that SH induces iWAT browning and prevents diet-induced obesity primarily through the AMPK/NRF2/HO-1 pathway by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Wenhui Liu
- Department of Endocrinology, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, China
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Huren Zou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Danming You
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Lingling Xu
- Department of Endocrinology, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, China
| |
Collapse
|
5
|
Zhang F, Fu Y, Wang J, Li F, Lang L, Zhu X, Wang L, Gao P, Shu G, Zhu C, Jiang Q, Wang S. Conjugated linoleic acid (CLA) reduces HFD-induced obesity by enhancing BAT thermogenesis and iWAT browning via the CD36-AMPK pathway. Cell Biochem Funct 2024; 42:e3937. [PMID: 38329451 DOI: 10.1002/cbf.3937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 02/09/2024]
Abstract
The antiobesity effect of conjugated linoleic acid (CLA) has been reported. However, the underlying mechanisms have not been fully clarified. Thus, this study aimed to investigate the effects of CLA on thermogenesis of interscapular brown adipose tissue (iBAT) and browning of inguinal subcutaneous white adipose tissue (iWAT) and explore the possible signaling pathway. The in vivo results showed that CLA enhanced the O2 consumption and heat production in HFD (high-fat diet)-fed female mice by roughly 38%. Meanwhile, CLA increased the average iBAT temperature by 2°C at the room temperature and cold exposure, respectively. Correspondingly, CLA caused 1.6- and 2.4-fold increases in the expression of UCP1 (uncoupling protein 1) of BAT and iWAT, respectively, suggesting the activated iBAT thermogenesis and iWAT browning in HFD-fed female mice. Meanwhile, CLA could promote the formation of brown and beige adipocytes in differentiated stromal vascular cells (SVCs) isolated from iBAT and iWAT (the expressions of UCP1 were promoted by about twofold changes). In possible mechanisms, CLA stimulated the expression of CD36 and the activation of the AMPK pathway in mice iBAT and iWAT as well as the differentiated SVCs. However, inhibition of CD36 and AMPK (adenosine 5'-monophosphate-activated protein kinase) abolished the promotive effects of CLA on brown and beige adipocytes formation. Hence, we showed that CLA reduced HFD-induced obesity through enhancing iBAT thermogenesis and iWAT browning via the CD36-AMPK pathway.
Collapse
Affiliation(s)
- Fenglin Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
| | - Yiming Fu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
| | - Junfeng Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
| | - Fan Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
| | - Limin Lang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
| | - Xiaotong Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
| | - Canjun Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, P. R. China
- Yunfu Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu, P. R. China
| |
Collapse
|
6
|
Abbasi K, Zarezadeh R, Valizadeh A, Mehdizadeh A, Hamishehkar H, Nouri M, Darabi M. White-brown adipose tissue interplay in polycystic ovary syndrome: Therapeutic avenues. Biochem Pharmacol 2024; 220:116012. [PMID: 38159686 DOI: 10.1016/j.bcp.2023.116012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
This study highlights the therapeutic potential of activating brown adipose tissue (BAT) for managing polycystic ovary syndrome (PCOS), a prevalent endocrine disorder associated with metabolic and reproductive abnormalities. BAT plays a crucial role in regulating energy expenditure and systemic insulin sensitivity, making it an attractive target for the treatment of obesity and metabolic diseases. Recent research suggests that impaired BAT function and mass may contribute to the link between metabolic disturbances and reproductive issues in PCOS. Additionally, abnormal white adipose tissue (WAT) can exacerbate these conditions by releasing adipokines and nonesterified fatty acids. In this review, we explored the impact of WAT changes on BAT function in PCOS and discussed the potential of BAT activation as a therapeutic strategy to improve PCOS symptoms. We propose that BAT activation holds promise for managing PCOS; however, further research is needed to confirm its efficacy and to develop clinically feasible methods for BAT activation.
Collapse
Affiliation(s)
- Khadijeh Abbasi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Valizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, Germany.
| |
Collapse
|
7
|
U-Din M, de Mello VD, Tuomainen M, Raiko J, Niemi T, Fromme T, Klåvus A, Gautier N, Haimilahti K, Lehtonen M, Kristiansen K, Newman JW, Pietiläinen KH, Pihlajamäki J, Amri EZ, Klingenspor M, Nuutila P, Pirinen E, Hanhineva K, Virtanen KA. Cold-stimulated brown adipose tissue activation is related to changes in serum metabolites relevant to NAD + metabolism in humans. Cell Rep 2023; 42:113131. [PMID: 37708023 DOI: 10.1016/j.celrep.2023.113131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/06/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023] Open
Abstract
Cold-induced brown adipose tissue (BAT) activation is considered to improve metabolic health. In murine BAT, cold increases the fundamental molecule for mitochondrial function, nicotinamide adenine dinucleotide (NAD+), but limited knowledge of NAD+ metabolism during cold in human BAT metabolism exists. We show that cold increases the serum metabolites of the NAD+ salvage pathway (nicotinamide and 1-methylnicotinamide) in humans. Additionally, individuals with cold-stimulated BAT activation have decreased levels of metabolites from the de novo NAD+ biosynthesis pathway (tryptophan, kynurenine). Serum nicotinamide correlates positively with cold-stimulated BAT activation, whereas tryptophan and kynurenine correlate negatively. Furthermore, the expression of genes involved in NAD+ biosynthesis in BAT is related to markers of metabolic health. Our data indicate that cold increases serum tryptophan conversion to nicotinamide to be further utilized by BAT. We conclude that NAD+ metabolism is activated upon cold in humans and is probably regulated in a coordinated fashion by several tissues.
Collapse
Affiliation(s)
- Mueez U-Din
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland
| | - Vanessa D de Mello
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Marjo Tuomainen
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Juho Raiko
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Tarja Niemi
- Department of Surgery, Turku University Hospital, Turku, Finland
| | - Tobias Fromme
- Chair for Molecular Nutritional Medicine, 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
| | - Anton Klåvus
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | | | - Kimmo Haimilahti
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; Research Program for Stem Cells and Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Marko Lehtonen
- Department of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | | | - John W Newman
- Obesity and Metabolism Research Unit, USDA-ARS Western Human Nutrition Research Center, Davis, CA, USA; West Coast Metabolomics Center, Davis Genome Center, University of California, Davis, Davis, CA 95616, USA; Department of Nutrition, University of California, Davis, Davis, CA 95616, USA
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Obesity Center, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Jussi Pihlajamäki
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Endocrinology and Clinical Nutrition, Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | | | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, 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
| | - Pirjo Nuutila
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland; Department of Endocrinology, Turku University Hospital, Turku, Finland
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; Research Unit for Internal Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Kati Hanhineva
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Life Technologies, Food Chemistry and Food Development Unit, University of Turku, Turku, Finland; Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Kirsi A Virtanen
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland; Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Endocrinology and Clinical Nutrition, Department of Medicine, Kuopio University Hospital, Kuopio, Finland; Department of Endocrinology, Turku University Hospital, Turku, Finland.
| |
Collapse
|
8
|
Noriega L, Yang CY, Wang CH. Brown Fat and Nutrition: Implications for Nutritional Interventions. Nutrients 2023; 15:4072. [PMID: 37764855 PMCID: PMC10536824 DOI: 10.3390/nu15184072] [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: 08/02/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Brown and beige adipocytes are renowned for their unique ability to generate heat through a mechanism known as thermogenesis. This process can be induced by exposure to cold, hormonal signals, drugs, and dietary factors. The activation of these thermogenic adipocytes holds promise for improving glucose metabolism, reducing fat accumulation, and enhancing insulin sensitivity. However, the translation of preclinical findings into effective clinical therapies poses challenges, warranting further research to identify the molecular mechanisms underlying the differentiation and function of brown and beige adipocytes. Consequently, research has focused on the development of drugs, such as mirabegron, ephedrine, and thyroid hormone, that mimic the effects of cold exposure to activate brown fat activity. Additionally, nutritional interventions have been explored as an alternative approach to minimize potential side effects. Brown fat and beige fat have emerged as promising targets for addressing nutritional imbalances, with the potential to develop strategies for mitigating the impact of metabolic diseases. Understanding the influence of nutritional factors on brown fat activity can facilitate the development of strategies to promote its activation and mitigate metabolic disorders.
Collapse
Affiliation(s)
- Lloyd Noriega
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Cheng-Ying Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Chih-Hao Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
- Graduate Institute of Cell Biology, College of Life Sciences, China Medical University, Taichung 406040, Taiwan
| |
Collapse
|
9
|
Jiang T, Su D, Liu X, Wang Y, Wang L. Transcriptomic Analysis Reveals Fibroblast Growth Factor 11 (FGF11) Role in Brown Adipocytes in Thermogenic Regulation of Goats. Int J Mol Sci 2023; 24:10838. [PMID: 37446019 DOI: 10.3390/ijms241310838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/18/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Brown adipose tissue (BAT) is the main site of adaptive thermogenesis, generates heat to maintain body temperature upon cold exposure, and protects against obesity by promoting energy expenditure. RNA-seq analysis revealed that FGF11 is enriched in BAT. However, the functions and regulatory mechanisms of FGF11 in BAT thermogenesis are still limited. In this study, we found that FGF11 was significantly enriched in goat BAT compared with white adipose tissue (WAT). Gain- and loss-of-function experiments revealed that FGF11 promoted differentiation and thermogenesis in brown adipocytes. However, FGF11 had no effect on white adipocyte differentiation. Furthermore, FGF11 promoted the expression of the UCP1 protein and an EBF2 element was responsible for UCP1 promoter activity. Additionally, FGF11 induced UCP1 gene expression through promoting EBF2 binding to the UCP1 promoter. These results revealed that FGF11 promotes differentiation and thermogenesis in brown adipocytes but not in white adipocytes of goats. These findings provide evidence for FGF11 and transcription factor regulatory functions in controlling brown adipose thermogenesis of goats.
Collapse
Affiliation(s)
- Tingting Jiang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Duo Su
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin Liu
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Wang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Linjie Wang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
10
|
Kong LR, Chen XH, Sun Q, Zhang KY, Xu L, Ding L, Zhou YP, Zhang ZB, Lin JR, Gao PJ. Loss of C3a and C5a receptors promotes adipocyte browning and attenuates diet-induced obesity via activating inosine/A2aR pathway. Cell Rep 2023; 42:112078. [PMID: 36735535 DOI: 10.1016/j.celrep.2023.112078] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/29/2022] [Accepted: 01/23/2023] [Indexed: 02/04/2023] Open
Abstract
Complement activation is thought to underline the pathologic progression of obesity-related metabolic disorders; however, its role in adaptive thermogenesis has scarcely been explored. Here, we identify complement C3a receptor (C3aR) and C5a receptor (C5aR) as critical switches to control adipocyte browning and energy balance in male mice. Loss of C3aR and C5aR in combination, more than individually, increases cold-induced adipocyte browning and attenuates diet-induced obesity in male mice. Mechanistically, loss of C3aR and C5aR increases regulatory T cell (Treg) accumulation in the subcutaneous white adipose tissue during cold exposure or high-fat diet. Activated Tregs produce adenosine, which is converted to inosine by adipocyte-derived adenosine deaminases. Inosine promotes adipocyte browning in a manner dependent on activating adenosine A2a receptor. These data reveal a regulatory mechanism of complement in controlling adaptive thermogenesis and suggest that targeting the C3aR/C5aR pathways may represent a therapeutic strategy in treating obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Ling-Ran Kong
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Hui Chen
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qing Sun
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai-Yuan Zhang
- Department of Traditional Chinese Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian Xu
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liliqiang Ding
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-Ping Zhou
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ze-Bei Zhang
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Rong Lin
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping-Jin Gao
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Shi J, Wang Q, Li C, Yang M, Hussain M, Zhang J, Feng F, Zhong H. Effects of the Novel LaPLa-Enriched Medium- and Long-Chain Triacylglycerols on Body Weight, Glycolipid Metabolism, and Gut Microbiota Composition in High Fat Diet-Fed C57BL/6J Mice. Molecules 2023; 28:molecules28020722. [PMID: 36677779 PMCID: PMC9861698 DOI: 10.3390/molecules28020722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
The roles of medium- and long-chain triacylglycerols (MLCT) on health benefits under high fat diet (HFD) conditions remain in dispute. This study was conducted to investigate the effects of novel LaPLa-rich MLCT on the glycolipid metabolism and gut microbiota in HFD-fed mice when pork fat is half replaced with MLCT and palm stearin (PS). The results showed that although MLCT could increase the body weight in the mouse model, it can improve the energy utilization, regulate the glucose and lipid metabolism, and inhibit the occurrence of inflammation. Furthermore, 16S rRNA gene sequencing of gut microbiota indicated that PS and MLCT affected the overall structure of the gut microbiota to a varying extent and specifically changed the abundance of some operational taxonomic units (OTUs). Moreover, several OTUs belonging to the genera Dorea, Streptococcus, and g_Eryipelotrichaceae had a high correlation with obesity and obesity-related metabolic disorders of the host. Therefore, it can be seen that this new MLCT has different properties and functions from the previous traditional MLCT, and it can better combine the advantages of MLCT, lauric acid, and sn-2 palmitate, as well as the advantages of health function and metabolism. In summary, this study explored the effects of LaPLa-enriched lipids on glycolipid metabolism in mice, providing theoretical support for future studies on the efficacy of different types of conjugated lipids, intending to apply them to industrial production and subsequent development of related products.
Collapse
Affiliation(s)
- Jinyuan Shi
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Qianqian Wang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Chuang Li
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Mengyu Yang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Muhammad Hussain
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Junhui Zhang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Fengqin Feng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
- Correspondence: (F.F.); (H.Z.); Tel.: +86-571-88982163 (F.F.); +86-571-88813585 (H.Z.)
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
- Correspondence: (F.F.); (H.Z.); Tel.: +86-571-88982163 (F.F.); +86-571-88813585 (H.Z.)
| |
Collapse
|
12
|
Yang J, Wang K, Yang Z. Treatment strategies for clear cell renal cell carcinoma: Past, present and future. Front Oncol 2023; 13:1133832. [PMID: 37025584 PMCID: PMC10070676 DOI: 10.3389/fonc.2023.1133832] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent histological subtype of kidney cancer, which is prone to metastasis, recurrence, and resistance to radiotherapy and chemotherapy. The burden it places on human health due to its refractory nature and rising incidence rate is substantial. Researchers have recently determined the ccRCC risk factors and optimized the clinical therapy based on the disease's underlying molecular mechanisms. In this paper, we review the established clinical therapies and novel potential therapeutic approaches for ccRCC, and we support the importance of investigating novel therapeutic options in the context of combining established therapies as a research hotspot, with the goal of providing diversified therapeutic options that promise to address the issue of drug resistance, with a view to the early realization of precision medicine and individualized treatment.
Collapse
Affiliation(s)
- Junwei Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Kuansong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhichun Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
- *Correspondence: Zhichun Yang,
| |
Collapse
|
13
|
Li L, Wan Q, Long Q, Nie T, Zhao S, Mao L, Cheng C, Zou C, Loomes K, Xu A, Lai L, Liu X, Duan Z, Hui X, Wu D. Comparative transcriptomic analysis of rabbit interscapular brown adipose tissue whitening under physiological conditions. Adipocyte 2022; 11:529-549. [PMID: 36000239 PMCID: PMC9427046 DOI: 10.1080/21623945.2022.2111053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 01/29/2023] Open
Abstract
Interscapular brown adipose tissue (iBAT) of both rabbits and humans exhibits a similar whitening phenomenon under physiological conditions. However, a detailed characterization of iBAT whitening in them is still lacking. Here, we chose rabbits as a model to gain a better understanding of the molecular signature changes during the whitening process of iBAT by transcriptomic analysis of rabbit iBAT at day 1, day 14, 1 month and 4 months after birth. We applied non-invasive MRI imaging to monitor the whitening process and correlated these changes with analysis of morphological, histological and molecular features. Principal component analysis (PCA) of differentially expressed genes delineated three major phases for the whitening process as Brown, Transition and Whitened BAT phases. RNA-sequencing data revealed that whitening of iBAT was an orchestrated process where multiple types of cells and tissues participated in a variety of physiological processes including neovascularization, formation of new nervous networks and immune regulation. Several key metabolic and signalling pathways contributed to whitening of iBAT, and immune cells and immune regulation appeared to play an overarching role.
Collapse
Affiliation(s)
- Lei Li
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qian Wan
- University of Chinese Academy of Sciences, Beijing, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qiaoyun Long
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong SAR
| | - Tao Nie
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shiting Zhao
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Liufeng Mao
- Clinical Department of Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Chuanli Cheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chao Zou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kerry Loomes
- School of Biological Sciences and Maurice Wilkins Centre, University of Auckland, New Zealand
| | - Aimin Xu
- Department of Medicine, University of Hong Kong, Hong Kong SAR
| | - Liangxue Lai
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ziyuan Duan
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Ziyuan Duan Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou510530, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaoyan Hui
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong SAR
- Xiaoyan Hui
| | - Donghai Wu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- CONTACT Donghai Wu
| |
Collapse
|
14
|
Wang J, Wu Q, Zhou Y, Yu L, Yu L, Deng Y, Tu C, Li W. The mechanisms underlying olanzapine-induced insulin resistance via the brown adipose tissue and the therapy in rats. Adipocyte 2022; 11:84-98. [PMID: 35067163 PMCID: PMC8786323 DOI: 10.1080/21623945.2022.2026590] [Citation(s) in RCA: 4] [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] [Indexed: 11/30/2022] Open
Abstract
A rapid increase has been observed in insulin resistance (IR) incidence induced by a long-term olanzapine treatment with no better ways to avoid it. Our study aimed to demonstrate the mechanism underlying the olanzapine-induced insulin resistance and find appropriate drug interventions. In this study, firstly, we constructed rat insulin resistance model using a two-month gavage of olanzapine and used the main active ingredient mixture of Gegen Qinlian Decoction for the treatment. The activity of brown adipose tissue (BAT) was measured using the PET/CT scan, whereas Western blot and quantitative real-time PCR were used to detect the expression of GLUT4 and UCP1. The results showed that the long-term administration of olanzapine impaired glucose tolerance and produced insulin resistance in rats, while Gegen Qinlian Decoction could improve this side effect. The results of the PET/CT scan showed that the BAT activity in the insulin-resistant rats was significantly lower than that of the Gegen Qinlian Decoction treated rats. Also, the expression of GLUT4 and UCP1 in the insulin resistance group showed a significant decrease, which could be up-regulated by Gegen Qinliane Decoction treatment. The results of both in vivo and in vitro experiments were consistent. we demonstrated that the olanzapine could induce IR in vitro and in vivo by decreasing the expression of UCP1; thus, suppressing the thermogenesis of BAT and impairing glucose uptake. More importantly, we demonstrated a possible novel strategy to improve the olanzapine-induced IR by Gegen Qinlian Decoction.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Zhou
- Department of Pharmacy, Wuhan Xirui Pharmaceutical Technology Co Ltd, Wuhan, China
| | - Liangyu Yu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lixiu Yu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yahui Deng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuyue Tu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiyong Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Tanaka R, Fuse-Hamaoka S, Kuroiwa M, Kurosawa Y, Endo T, Kime R, Yoneshiro T, Hamaoka T. The Effects of 10-Week Strength Training in the Winter on Brown-like Adipose Tissue Vascular Density. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10375. [PMID: 36012011 PMCID: PMC9408462 DOI: 10.3390/ijerph191610375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
There is no evidence of the effect of exercise training on human brown-like adipose tissue vascular density (BAT-d). Here, we report whether whole-body strength training (ST) in a cold environment increased BAT-d. The participants were 18 men aged 20-31 years. They were randomly assigned to two groups: one that performed ST twice a week at 75% intensity of one-repetition maximum for 10 weeks during winter (EX; n = 9) and a control group that did not perform ST (CT; n = 9). The total hemoglobin concentration in the supraclavicular region determined by time-resolved near-infrared spectroscopy was used as a parameter of BAT-d. ST volume (Tvol) was defined as the mean of the weight × repetition × sets of seven training movements. The number of occasions where the room temperature was lower than the median (NRcold) was counted as an index of potential cold exposure during ST. There was no significant between-group difference in BAT-d. Multiple regression analysis using body mass index, body fat percentage, NRcold, and Tvol as independent variables revealed that NRcold and Tvol were determined as predictive of changes in BAT-d. An appropriate combination of ST with cold environments could be an effective strategy for modulating BAT.
Collapse
Affiliation(s)
- Riki Tanaka
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Sayuri Fuse-Hamaoka
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Miyuki Kuroiwa
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Yuko Kurosawa
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Tasuki Endo
- Faculty of Science and Technology, Meijo University, Nagoya 468-8502, Japan
| | - Ryotaro Kime
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Takeshi Yoneshiro
- Research Center for Advanced Science and Technology, University of Tokyo, Tokyo 153-8904, Japan
| | - Takafumi Hamaoka
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|
16
|
Scheele C, Henriksen TI, Nielsen S. Isolation and Characterization of Human Brown Adipocytes. Methods Mol Biol 2022; 2448:217-234. [PMID: 35167100 DOI: 10.1007/978-1-0716-2087-8_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Brown adipose tissue (BAT) is a thermoregulatory fat with energy-consuming properties. The location and heterogeneity of this tissue makes it complicated to sample before and after interventions in humans, and an in vitro model for mechanistic and molecular studies is therefore of great value. We here describe a protocol for isolation of progenitors from the stromal vascular fraction of BAT biopsies obtained surgically from adult humans. We further present how these cells are differentiated in vitro and finally how they are characterized for thermogenic capacity. Methods for characterization described here include norepinephrine-induced thermogenic gene expression using qPCR; norepinephrine-induced mitochondrial uncoupling using the Seahorse XFe96 Analyzer, and norepinephrine-induced expression of UCP1 using the RNAscope® Technology.
Collapse
Affiliation(s)
- Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| | - Tora Ida Henriksen
- Centre for Physical Activity Research, Department of Infectious Diseases, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Nielsen
- Centre for Physical Activity Research, Department of Infectious Diseases, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Mukherjee R, Sanchez-Gurmaches J. Fluorescent Genetic Tools for Studying Brown Fat Development and Function in Mice. Methods Mol Biol 2022; 2448:203-215. [PMID: 35167099 PMCID: PMC10112487 DOI: 10.1007/978-1-0716-2087-8_13] [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] [Indexed: 10/19/2022]
Abstract
Techniques to trace and isolate brown adipocyte precursor and adipocytes during development and disease are essential to fully understand brown adipose tissue development and function. Here we report several protocols using the R26R-mTmG reporter mice in thermogenic tissues based on confocal microscopy and fluorescence based flow cytometry. These techniques may be useful to understand the influence of genetic or environmental alterations in brown adipocyte precursors and adipocyte biology.
Collapse
Affiliation(s)
- Rajib Mukherjee
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joan Sanchez-Gurmaches
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
18
|
Xu XY, Zhao CN, Li BY, Tang GY, Shang A, Gan RY, Feng YB, Li HB. Effects and mechanisms of tea on obesity. Crit Rev Food Sci Nutr 2021:1-18. [PMID: 34704503 DOI: 10.1080/10408398.2021.1992748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Obesity has become a global health concern. It increases the risk of several diseases, such as type 2 diabetes mellitus, nonalcoholic fatty liver disease, and certain cancers, which threatens human health and increases social economic burden. As one of the most consumed beverages, tea contains various phytochemicals with potent bioactive properties and health-promoting effects, such as antioxidant, immune-regulation, cardiovascular protection and anticancer. Tea and its components are also considered as potential candidates for anti-obesity. Epidemiological studies indicate that regular consumption of tea is beneficial for reducing body fat. In addition, the experimental studies demonstrate that the potential anti-obesity mechanisms of tea are mainly involved in increasing energy expenditure and lipid catabolism, decreasing nutrient digestion and absorption as well as lipid synthesis, and regulating adipocytes, neuroendocrine system and gut microbiota. Moreover, most of clinical studies illustrate that the intake of green tea could reduce body weight and alleviate the obesity. In this review, we focus on the effect of tea and its components on obesity from epidemiological, experimental, and clinical studies, and discuss their potential mechanisms.
Collapse
Affiliation(s)
- Xiao-Yu Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China.,Li Ka Shing Faculty of Medicine, School of Chinese Medicine, The University of Hong Kong, China Hong Kong
| | - Cai-Ning Zhao
- Li Ka Shing Faculty of Medicine, Department of Clinical Oncology, The University of Hong Kong, China Hong Kong
| | - Bang-Yan Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Guo-Yi Tang
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, The University of Hong Kong, China Hong Kong
| | - Ao Shang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China.,Li Ka Shing Faculty of Medicine, School of Chinese Medicine, The University of Hong Kong, China Hong Kong
| | - Ren-You Gan
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China.,Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Yi-Bin Feng
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, The University of Hong Kong, China Hong Kong
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
19
|
Pescador N, Francisco V, Vázquez P, Esquinas EM, González-Páramos C, Valdecantos MP, García-Martínez I, Urrutia AA, Ruiz L, Escalona-Garrido C, Foretz M, Viollet B, Fernández-Moreno MÁ, Calle-Pascual AL, Obregón MJ, Aragonés J, Valverde ÁM. Metformin reduces macrophage HIF1α-dependent proinflammatory signaling to restore brown adipocyte function in vitro. Redox Biol 2021; 48:102171. [PMID: 34736121 PMCID: PMC8577482 DOI: 10.1016/j.redox.2021.102171] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 12/25/2022] Open
Abstract
Therapeutic potential of metformin in obese/diabetic patients has been associated to its ability to combat insulin resistance. However, it remains largely unknown the signaling pathways involved and whether some cell types are particularly relevant for its beneficial effects. M1-activation of macrophages by bacterial lipopolysaccharide (LPS) promotes a paracrine activation of hypoxia-inducible factor-1α (HIF1α) in brown adipocytes which reduces insulin signaling and glucose uptake, as well as β-adrenergic sensitivity. Addition of metformin to M1-polarized macrophages blunted these signs of brown adipocyte dysfunction. At the molecular level, metformin inhibits an inflammatory program executed by HIF1α in macrophages by inducing its degradation through the inhibition of mitochondrial complex I activity, thereby reducing oxygen consumption in a reactive oxygen species (ROS)-independent manner. In obese mice, metformin reduced inflammatory features in brown adipose tissue (BAT) such as macrophage infiltration, proinflammatory signaling and gene expression, and restored the response to cold exposure. In conclusion, the impact of metformin on macrophages by suppressing a HIF1α-dependent proinflammatory program is likely responsible for a secondary beneficial effect on insulin-mediated glucose uptake and β-adrenergic responses in brown adipocytes.
Collapse
Affiliation(s)
- Nuria Pescador
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain.
| | - Vera Francisco
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Patricia Vázquez
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Eva María Esquinas
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Cristina González-Páramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Departamento de Bioquímica. Facultad de Medicina. Universidad Autónoma de Madrid, Spain and Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Instituto de Salud Carlos III, Madrid, Spain
| | - M Pilar Valdecantos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Irma García-Martínez
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Andrés A Urrutia
- Research Unit, Hospital de La Princesa, Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Spain
| | - Laura Ruiz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Escalona-Garrido
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Marc Foretz
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Benoit Viollet
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Miguel Ángel Fernández-Moreno
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Departamento de Bioquímica. Facultad de Medicina. Universidad Autónoma de Madrid, Spain and Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso L Calle-Pascual
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Endocrinología y Nutrición, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria Del Hospital Clínico San Carlos (IdISSC), Madrid, Spain; Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - María Jesús Obregón
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Julián Aragonés
- Research Unit, Hospital de La Princesa, Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERcv), Instituto de Salud Carlos III, Madrid, Spain
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
20
|
Cavalcanti-de-Albuquerque JP, Donato J. Rolling out physical exercise and energy homeostasis: Focus on hypothalamic circuitries. Front Neuroendocrinol 2021; 63:100944. [PMID: 34425188 DOI: 10.1016/j.yfrne.2021.100944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 01/17/2023]
Abstract
Energy balance is the fine regulation of energy expenditure and energy intake. Negative energy balance causes body weight loss, while positive energy balance promotes weight gain. Modern societies offer a maladapted way of life, where easy access to palatable foods and the lack of opportunities to perform physical activity are considered the roots of the obesity pandemic. Physical exercise increases energy expenditure and, consequently, is supposed to promote weight loss. Paradoxically, physical exercise acutely drives anorexigenic-like effects, but the mechanisms are still poorly understood. Using an evolutionary background, this review aims to highlight the potential involvement of the melanocortin system and other hypothalamic neural circuitries regulating energy balance during and after physical exercise. The physiological significance of these changes will be explored, and possible signalling agents will be addressed. The knowledge discussed here might be important for clarifying obesity aetiology as well as new therapeutic approaches for body weight loss.
Collapse
Affiliation(s)
| | - José Donato
- Department of Physiology and Biophysics, University of São Paulo, São Paulo 05508-900, Brazil.
| |
Collapse
|
21
|
Saitoh S, Van Wijk K, Nakajima O. Crosstalk between Metabolic Disorders and Immune Cells. Int J Mol Sci 2021; 22:ijms221810017. [PMID: 34576181 PMCID: PMC8469678 DOI: 10.3390/ijms221810017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 12/22/2022] Open
Abstract
Metabolic syndrome results from multiple risk factors that arise from insulin resistance induced by abnormal fat deposition. Chronic inflammation owing to obesity primarily results from the recruitment of pro-inflammatory M1 macrophages into the adipose tissue stroma, as the adipocytes within become hypertrophied. During obesity-induced inflammation in adipose tissue, pro-inflammatory cytokines are produced by macrophages and recruit further pro-inflammatory immune cells into the adipose tissue to boost the immune response. Here, we provide an overview of the biology of macrophages in adipose tissue and the relationship between other immune cells, such as CD4+ T cells, natural killer cells, and innate lymphoid cells, and obesity and type 2 diabetes. Finally, we discuss the link between the human pathology and immune response and metabolism and further highlight potential therapeutic targets for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Shinichi Saitoh
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan;
| | - Koen Van Wijk
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan;
| | - Osamu Nakajima
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan;
- Correspondence:
| |
Collapse
|
22
|
Nazeri A, Crandall JP, Fraum TJ, Wahl RL. Repeatability of Radiomic Features of Brown Adipose Tissue. J Nucl Med 2021; 62:700-706. [PMID: 33037091 DOI: 10.2967/jnumed.120.248674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to assess the repeatability of activated brown adipose tissue (BAT) radiomic features. To decipher radiomic features that may provide useful information on BAT, the impact of reconstruction methods and imaging modality choice was also evaluated. Methods: Twenty-seven healthy adults enrolled in this study. After a cooling procedure to activate BAT, volunteers underwent 18F-FDG imaging. Participants underwent repeat imaging using the same imaging protocols and a similar 18F-FDG dose within 14 d. Active BAT was segmented using the BARCIST 1.0 methods. Radiomic features were extracted from each region of interest on high-definition PET (HD PET), non-HD PET, and CT images. Lin's concordance correlation coefficient was used to estimate the repeatability of the extracted radiomic features. To determine whether BAT radiomic feature repeatability correlates with BAT SUVmax repeatability, participants were stratified based on the relative difference in SUVmax between sessions. Non-HD PET repeatable features were clustered together using hierarchical clustering, and the normalized dynamic range of each feature was computed to identify the most informative feature within each cluster. Results: Eighteen of the 27 volunteers had sufficient BAT activity for radiomic analysis. Sixty-six HD PET, 66 non-HD PET, and 6 CT features showed high repeatability (concordance correlation coefficient ≥ 0.80). Feature repeatability was significantly higher for PET than for CT, but there was no statistically significant difference between HD and non-HD PET in radiomic feature repeatability. The repeatability of radiomic features extracted from each modality and reconstruction method type followed the trend in SUVmax, as participants with lower relative differences in SUVmax between initial and repeated imaging sessions had higher radiomic feature repeatability. Hierarchical clustering of the high-repeatability PET features resulted in 10 highly correlated clusters (R 2 ≥ 0.95). Seven features, including SUVmax, did not cluster with any other features. Conclusion: Several clusters of highly repeatable BAT radiomic features derived from 18F-FDG PET/CT appear to provide information regarding BAT activity distinct from SUVmax These features might be explored as quantitative imaging biomarkers of BAT activity in future studies.
Collapse
Affiliation(s)
- Aria Nazeri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - John P Crandall
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Tyler J Fraum
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Richard L Wahl
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
23
|
Verduci E, Calcaterra V, Di Profio E, Fiore G, Rey F, Magenes VC, Todisco CF, Carelli S, Zuccotti GV. Brown Adipose Tissue: New Challenges for Prevention of Childhood Obesity. A Narrative Review. Nutrients 2021; 13:nu13051450. [PMID: 33923364 PMCID: PMC8145569 DOI: 10.3390/nu13051450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/14/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Pediatric obesity remains a challenge in modern society. Recently, research has focused on the role of the brown adipose tissue (BAT) as a potential target of intervention. In this review, we revised preclinical and clinical works on factors that may promote BAT or browning of white adipose tissue (WAT) from fetal age to adolescence. Maternal lifestyle, type of breastfeeding and healthy microbiota can affect the thermogenic activity of BAT. Environmental factors such as exposure to cold or physical activity also play a role in promoting and activating BAT. Most of the evidence is preclinical, although in clinic there is some evidence on the role of omega-3 PUFAs (EPA and DHA) supplementation on BAT activation. Clinical studies are needed to dissect the early factors and their modulation to allow proper BAT development and functions and to prevent onset of childhood obesity.
Collapse
Affiliation(s)
- Elvira Verduci
- Department of Health Sciences, University of Milan, 20146 Milan, Italy
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Correspondence: (E.V.); (S.C.)
| | - Valeria Calcaterra
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| | - Elisabetta Di Profio
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Department of Animal Sciences for Health, Animal Production and Food Safety, University of Milan, 20133 Milan, Italy
| | - Giulia Fiore
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Federica Rey
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
| | - Vittoria Carlotta Magenes
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Carolina Federica Todisco
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
- Correspondence: (E.V.); (S.C.)
| | - Gian Vincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
| |
Collapse
|
24
|
Felder M, Maushart CI, Gashi G, Senn JR, Becker AS, Müller J, Balaz M, Wolfrum C, Burger IA, Betz MJ. Fluvastatin Reduces Glucose Tolerance in Healthy Young Individuals Independently of Cold Induced BAT Activity. Front Endocrinol (Lausanne) 2021; 12:765807. [PMID: 34858338 PMCID: PMC8631514 DOI: 10.3389/fendo.2021.765807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/14/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Statins are commonly prescribed for primary and secondary prevention of atherosclerotic disease. They reduce cholesterol biosynthesis by inhibiting hydroxymethylglutaryl-coenzyme A-reductase (HMG-CoA-reductase) and therefore mevalonate synthesis. Several studies reported a small, but significant increase in the diagnosis of diabetes mellitus with statin treatment. The molecular mechanisms behind this adverse effect are not yet fully understood. Brown adipose tissue (BAT), which plays a role in thermogenesis, has been associated with a reduced risk of insulin resistance. Statins inhibit adipose tissue browning and have been negatively linked to the presence of BAT in humans. We therefore speculated that inhibition of BAT by statins contributes to increased insulin resistance in humans. METHODS A prospective study was conducted in 17 young, healthy men. After screening whether significant cold-induced thermogenesis (CIT) was present, participants underwent glucose tolerance testing (oGTT) and assessment of BAT activity by FDG-PET/MRI after cold-exposure and treatment with a β3-agonist. Fluvastatin 2x40mg per day was then administered for two weeks and oGTT and FDG-PET/MRI were repeated. RESULTS Two weeks of fluvastatin treatment led to a significant increase in glucose area under the curve (AUC) during oGTT (p=0.02), reduction in total cholesterol and LDL cholesterol (both p<0.0001). Insulin AUC (p=0.26), resting energy expenditure (REE) (p=0.44) and diet induced thermogenesis (DIT) (p=0.27) did not change significantly. The Matsuda index, as an indicator of insulin sensitivity, was lower after fluvastatin intake, but the difference was not statistically significant (p=0.09). As parameters of BAT activity, mean standard uptake value (SUVmean) (p=0.12), volume (p=0.49) and total glycolysis (p=0.74) did not change significantly during the intervention. Matsuda index, was inversely related to SUVmean and the respiratory exchange ratio (RER) (both R2 = 0.44, p=0.005) at baseline, but not after administration of fluvastatin (R2 = 0.08, p=0.29, and R2 = 0.14, p=0.16, respectively). CONCLUSIONS Treatment with fluvastatin for two weeks reduced serum lipid levels but increased glucose AUC in young, healthy men, indicating reduced glucose tolerance. This was not associated with changes in cold-induced BAT activity.
Collapse
Affiliation(s)
- Martina Felder
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Claudia Irene Maushart
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Gani Gashi
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jaël Rut Senn
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Anton S. Becker
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich/University of Zurich, Zurich, Switzerland
| | - Julian Müller
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich/University of Zurich, Zurich, Switzerland
| | - Miroslav Balaz
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Irene A. Burger
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich/University of Zurich, Zurich, Switzerland
| | - Matthias Johannes Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
- *Correspondence: Matthias Johannes Betz,
| |
Collapse
|
25
|
Rabiee A, Plucińska K, Isidor MS, Brown EL, Tozzi M, Sidoli S, Petersen PSS, Agueda-Oyarzabal M, Torsetnes SB, Chehabi GN, Lundh M, Altıntaş A, Barrès R, Jensen ON, Gerhart-Hines Z, Emanuelli B. White adipose remodeling during browning in mice involves YBX1 to drive thermogenic commitment. Mol Metab 2020; 44:101137. [PMID: 33285300 PMCID: PMC7779825 DOI: 10.1016/j.molmet.2020.101137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/16/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Objective Increasing adaptive thermogenesis by stimulating browning in white adipose tissue is a promising method of improving metabolic health. However, the molecular mechanisms underlying this transition remain elusive. Our study examined the molecular determinants driving the differentiation of precursor cells into thermogenic adipocytes. Methods In this study, we conducted temporal high-resolution proteomic analysis of subcutaneous white adipose tissue (scWAT) after cold exposure in mice. This was followed by loss- and gain-of-function experiments using siRNA-mediated knockdown and CRISPRa-mediated induction of gene expression, respectively, to evaluate the function of the transcriptional regulator Y box-binding protein 1 (YBX1) during adipogenesis of brown pre-adipocytes and mesenchymal stem cells. Transcriptomic analysis of mesenchymal stem cells following induction of endogenous Ybx1 expression was conducted to elucidate transcriptomic events controlled by YBX1 during adipogenesis. Results Our proteomics analysis uncovered 509 proteins differentially regulated by cold in a time-dependent manner. Overall, 44 transcriptional regulators were acutely upregulated following cold exposure, among which included the cold-shock domain containing protein YBX1, peaking after 24 h. Cold-induced upregulation of YBX1 also occurred in brown adipose tissue, but not in visceral white adipose tissue, suggesting a role of YBX1 in thermogenesis. This role was confirmed by Ybx1 knockdown in brown and brite preadipocytes, which significantly impaired their thermogenic potential. Conversely, inducing Ybx1 expression in mesenchymal stem cells during adipogenesis promoted browning concurrent with an increased expression of thermogenic markers and enhanced mitochondrial respiration. At a molecular level, our transcriptomic analysis showed that YBX1 regulates a subset of genes, including the histone H3K9 demethylase Jmjd1c, to promote thermogenic adipocyte differentiation. Conclusion Our study mapped the dynamic proteomic changes of murine scWAT during browning and identified YBX1 as a novel factor coordinating the genomic mechanisms by which preadipocytes commit to brite/beige lineage. Dynamic proteome remodeling occurs in mouse subcutaneous white fat with cold. YBX1 acutely increases in response to cold in thermogenic adipose tissues. YBX1 is required for the optimal implementation of the early thermogenic program. YBX1 promotes metabolic and thermogenic programs and enhances mitochondrial respiration.
Collapse
Affiliation(s)
- Atefeh Rabiee
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kaja Plucińska
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie Sophie Isidor
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Erin Louise Brown
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marco Tozzi
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, Denmark
| | - Patricia Stephanie S Petersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marina Agueda-Oyarzabal
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Silje Bøen Torsetnes
- Department of Biochemistry and Molecular Biology and VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Galal Nazih Chehabi
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Lundh
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ali Altıntaş
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ole Nørregaard Jensen
- Department of Biochemistry and Molecular Biology and VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Brice Emanuelli
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
26
|
Abstract
Since the discovery of functionally competent, energy-consuming brown adipose tissue (BAT) in adult humans, much effort has been devoted to exploring this tissue as a means for increasing energy expenditure to counteract obesity. However, despite promising effects on metabolic rate and insulin sensitivity, no convincing evidence for weight-loss effects of cold-activated human BAT exists to date. Indeed, increasing energy expenditure would naturally induce compensatory feedback mechanisms to defend body weight. Interestingly, BAT is regulated by multiple interactions with the hypothalamus from regions overlapping with centers for feeding behavior and metabolic control. Therefore, in the further exploration of BAT as a potential source of novel drug targets, we discuss the hypothalamic orchestration of BAT activity and the relatively unexplored BAT feedback mechanisms on neuronal regulation. With a holistic view on hypothalamic-BAT interactions, we aim to raise ideas and provide a new perspective on this circuit and highlight its clinical relevance.
Collapse
Affiliation(s)
- Jo B Henningsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark;
| |
Collapse
|
27
|
Li Y, Schwalie PC, Bast-Habersbrunner A, Mocek S, Russeil J, Fromme T, Deplancke B, Klingenspor M. Systems-Genetics-Based Inference of a Core Regulatory Network Underlying White Fat Browning. Cell Rep 2020; 29:4099-4113.e5. [PMID: 31851936 DOI: 10.1016/j.celrep.2019.11.053] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/02/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
Recruitment of brite/beige cells, known as browning of white adipose tissue (WAT), is an efficient way to turn an energy-storing organ into an energy-dissipating one and may therefore be of therapeutic value in combating obesity. However, a comprehensive understanding of the regulatory mechanisms mediating WAT browning is still lacking. Here, we exploit the large natural variation in WAT browning propensity between inbred mouse strains to gain an inclusive view of the core regulatory network coordinating this cellular process. Combining comparative transcriptomics, perturbation-based validations, and gene network analyses, we present a comprehensive gene regulatory network of inguinal WAT browning, revealing up to four distinct regulatory modules with key roles for uncovered transcriptional factors, while also providing deep insights into the genetic architecture of brite adipogenesis. The presented findings therefore greatly increase our understanding of the molecular drivers mediating the intriguing cellular heterogeneity and plasticity of adipose tissue.
Collapse
Affiliation(s)
- Yongguo Li
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; EKFZ-Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Petra C Schwalie
- Institute of Bio-engineering, School of Life Sciences, EPFL and Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Andrea Bast-Habersbrunner
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; EKFZ-Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Sabine Mocek
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; EKFZ-Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Julie Russeil
- Institute of Bio-engineering, School of Life Sciences, EPFL and Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Tobias Fromme
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; EKFZ-Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Bart Deplancke
- Institute of Bio-engineering, School of Life Sciences, EPFL and Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland.
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; EKFZ-Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany.
| |
Collapse
|
28
|
Zhou Z, Moore TM, Drew BG, Ribas V, Wanagat J, Civelek M, Segawa M, Wolf DM, Norheim F, Seldin MM, Strumwasser AR, Whitney KA, Lester E, Reddish BR, Vergnes L, Reue K, Rajbhandari P, Tontonoz P, Lee J, Mahata SK, Hewitt SC, Shirihai O, Gastonbury C, Small KS, Laakso M, Jensen J, Lee S, Drevon CA, Korach KS, Lusis AJ, Hevener AL. Estrogen receptor α controls metabolism in white and brown adipocytes by regulating Polg1 and mitochondrial remodeling. Sci Transl Med 2020; 12:eaax8096. [PMID: 32759275 PMCID: PMC8212422 DOI: 10.1126/scitranslmed.aax8096] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 11/04/2019] [Accepted: 07/16/2020] [Indexed: 12/14/2022]
Abstract
Obesity is heightened during aging, and although the estrogen receptor α (ERα) has been implicated in the prevention of obesity, its molecular actions in adipocytes remain inadequately understood. Here, we show that adipose tissue ESR1/Esr1 expression inversely associated with adiposity and positively associated with genes involved in mitochondrial metabolism and markers of metabolic health in 700 Finnish men and 100 strains of inbred mice from the UCLA Hybrid Mouse Diversity Panel. To determine the anti-obesity actions of ERα in fat, we selectively deleted Esr1 from white and brown adipocytes in mice. In white adipose tissue, Esr1 controlled oxidative metabolism by restraining the targeted elimination of mitochondria via the E3 ubiquitin ligase parkin. mtDNA content was elevated, and adipose tissue mass was reduced in adipose-selective parkin knockout mice. In brown fat centrally involved in body temperature maintenance, Esr1 was requisite for both mitochondrial remodeling by dynamin-related protein 1 (Drp1) and uncoupled respiration thermogenesis by uncoupled protein 1 (Ucp1). In both white and brown fat of female mice and adipocytes in culture, mitochondrial dysfunction in the context of Esr1 deletion was paralleled by a reduction in the expression of the mtDNA polymerase γ subunit Polg1 We identified Polg1 as an ERα target gene by showing that ERα binds the Polg1 promoter to control its expression in 3T3L1 adipocytes. These findings support strategies leveraging ERα action on mitochondrial function in adipocytes to combat obesity and metabolic dysfunction.
Collapse
Affiliation(s)
- Zhenqi Zhou
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Timothy M Moore
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Brian G Drew
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Vicent Ribas
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Jonathan Wanagat
- Division of Geriatrics, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Mete Civelek
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Mayuko Segawa
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Dane M Wolf
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Frode Norheim
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Marcus M Seldin
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Alexander R Strumwasser
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Kate A Whitney
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Ellen Lester
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Britany R Reddish
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Laurent Vergnes
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Prashant Rajbhandari
- Department of Pathology and Laboratory Medicine and the Howard Hughes Research Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine and the Howard Hughes Research Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Jason Lee
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Sushil K Mahata
- VA San Diego Healthcare System, San Diego, CA 92161, USA
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Sylvia C Hewitt
- Receptor Biology Section, NIEHS, NIH, Research Triangle Park, NC 27709, USA
| | - Orian Shirihai
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Craig Gastonbury
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE17EH, UK
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE17EH, UK
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio 70210, Finland
| | - Jorgen Jensen
- Department of Physical Performance, Norwegian School of Sport Science, Oslo 0806, Norway
| | - Sindre Lee
- University Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo 0316, Norway
| | - Christian A Drevon
- University Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo 0316, Norway
| | - Kenneth S Korach
- Receptor Biology Section, NIEHS, NIH, Research Triangle Park, NC 27709, USA
| | - Aldons J Lusis
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Andrea L Hevener
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA.
- Iris Cantor-UCLA Women's Health Research Center, Los Angeles, CA 90095, USA
| |
Collapse
|
29
|
Vonhögen IG, el Azzouzi H, Olieslagers S, Vasilevich A, de Boer J, Tinahones FJ, da Costa Martins PA, de Windt LJ, Murri M. MiR-337-3p Promotes Adipocyte Browning by Inhibiting TWIST1. Cells 2020; 9:cells9041056. [PMID: 32340411 PMCID: PMC7226112 DOI: 10.3390/cells9041056] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 01/04/2023] Open
Abstract
The prevalence of metabolic syndrome (MetS) and obesity is an alarming health issue worldwide. Obesity is characterized by an excessive accumulation of white adipose tissue (WAT), and it is associated with diminished brown adipose tissue (BAT) activity. Twist1 acts as a negative feedback regulator of BAT metabolism. Therefore, targeting Twist1 could become a strategy for obesity and metabolic disease. Here, we have identified miR-337-3p as an upstream regulator of Twist1. Increased miR-337-3p expression paralleled decreased expression of TWIST1 in BAT compared to WAT. Overexpression of miR-337-3p in brown pre-adipocytes provoked a reduction in Twist1 expression that was accompanied by increased expression of brown/mitochondrial markers. Luciferase assays confirmed an interaction between the miR-337 seed sequence and Twist1 3′UTR. The inverse relationship between the expression of TWIST1 and miR-337 was finally validated in adipose tissue samples from non-MetS and MetS subjects that demonstrated a dysregulation of the miR-337-Twist1 molecular axis in MetS. The present study demonstrates that adipocyte miR-337-3p suppresses Twist1 repression and enhances the browning of adipocytes.
Collapse
Affiliation(s)
- Indira G.C. Vonhögen
- Department of Molecular Genetics, Faculty of Sciences and Engineering, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands; (I.G.C.V.); (H.e.A.); (S.O.); (P.A.d.C.M.)
| | - Hamid el Azzouzi
- Department of Molecular Genetics, Faculty of Sciences and Engineering, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands; (I.G.C.V.); (H.e.A.); (S.O.); (P.A.d.C.M.)
- Department of Molecular Genetics, Erasmus University MC, 3015 GD Rotterdam, The Netherlands
| | - Servé Olieslagers
- Department of Molecular Genetics, Faculty of Sciences and Engineering, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands; (I.G.C.V.); (H.e.A.); (S.O.); (P.A.d.C.M.)
| | - Aliaksei Vasilevich
- BioInterface Science Group, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands; (A.V.); (J.d.B.)
| | - Jan de Boer
- BioInterface Science Group, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands; (A.V.); (J.d.B.)
| | - Francisco J. Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (F.J.T.); (M.M.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERObn, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Faculty of Medicine, University of Malaga, 29010 Malaga, Spain
| | - Paula A. da Costa Martins
- Department of Molecular Genetics, Faculty of Sciences and Engineering, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands; (I.G.C.V.); (H.e.A.); (S.O.); (P.A.d.C.M.)
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Leon J. de Windt
- Department of Molecular Genetics, Faculty of Sciences and Engineering, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands; (I.G.C.V.); (H.e.A.); (S.O.); (P.A.d.C.M.)
- Correspondence:
| | - Mora Murri
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (F.J.T.); (M.M.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERObn, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
30
|
Tanaka R, Fuse S, Kuroiwa M, Amagasa S, Endo T, Ando A, Kime R, Kurosawa Y, Hamaoka T. Vigorous-Intensity Physical Activities Are Associated with High Brown Adipose Tissue Density in Humans. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E2796. [PMID: 32325644 PMCID: PMC7216014 DOI: 10.3390/ijerph17082796] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 12/31/2022]
Abstract
Brown adipose tissue (BAT) plays a role in adaptive thermogenesis in response to cold environments and dietary intake via sympathetic nervous system (SNS) activation. It is unclear whether physical activity increases BAT density (BAT-d). Two-hundred ninety-eight participants (age: 41.2 ± 12.1 (mean ± standard deviation), height: 163.6 ± 8.3 cm, weight: 60.2 ± 11.0 kg, body mass index (BMI): 22.4 ± 3.0 kg/m2, body fat percentage: 25.4 ± 7.5%) without smoking habits were categorized based on their physical activity levels (a group performing physical activities including walking and moderate physical activity (WM) and a group performing WM + vigorous-intensity physical activities (VWM)). We measured the total hemoglobin concentration ([Total-Hb]) in the supraclavicular region, an index of BAT-d, and anthropometric parameters. [Total-Hb] was significantly higher in VWM than WM for all participant groups presumably owing to SNS activation during vigorous-intensity physical activities, and unrelated to the amount of total physical activity levels. Furthermore, multiple regression analysis revealed that BAT-d was related to visceral fat area and VWM in men and related to body fat percentage in women. We conclude that vigorous-intensity physical activities are associated with high BAT-d in humans, especially in men.
Collapse
Affiliation(s)
- Riki Tanaka
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan; (R.T.); (S.F.); (M.K.); (T.E.); (R.K.); (Y.K.)
| | - Sayuri Fuse
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan; (R.T.); (S.F.); (M.K.); (T.E.); (R.K.); (Y.K.)
| | - Miyuki Kuroiwa
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan; (R.T.); (S.F.); (M.K.); (T.E.); (R.K.); (Y.K.)
| | - Shiho Amagasa
- Department of Preventive Medicine and Public Health, Tokyo Medical University, Tokyo 160-8402, Japan;
| | - Tasuki Endo
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan; (R.T.); (S.F.); (M.K.); (T.E.); (R.K.); (Y.K.)
| | - Akira Ando
- Japan Institute of Sports Sciences, Tokyo 115-0056, Japan;
| | - Ryotaro Kime
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan; (R.T.); (S.F.); (M.K.); (T.E.); (R.K.); (Y.K.)
| | - Yuko Kurosawa
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan; (R.T.); (S.F.); (M.K.); (T.E.); (R.K.); (Y.K.)
| | - Takafumi Hamaoka
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Tokyo 160-8402, Japan; (R.T.); (S.F.); (M.K.); (T.E.); (R.K.); (Y.K.)
| |
Collapse
|
31
|
Liu M, Yun P, Hu Y, Yang J, Khadka RB, Peng X. Effects of Grape Seed Proanthocyanidin Extract on Obesity. Obes Facts 2020; 13:279-291. [PMID: 32114568 PMCID: PMC7250358 DOI: 10.1159/000502235] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/19/2019] [Indexed: 12/11/2022] Open
Abstract
Obesity is a chronic metabolic disease resulting from excessive fat accumulation and/or abnormal distribution caused by multiple factors. As a major component of metabolic syndrome, obesity is closely related to many diseases such as type 2 diabetes mellitus, hyperlipidemia, hypertension, coronary heart disease, stroke and cancer. Hence, the problem of obesity cannot be ignored, and recent studies have shown that grape seed proanthocyanidin extract (GSPE) has an antiobesity effect. This paper systematically reviews the research progress and potential mechanism of GSPE emphasizing on obesity prevention and treatment.
Collapse
Affiliation(s)
- Miao Liu
- Medical School of Yangtze University, Jingzhou, China
| | - Peng Yun
- Department of Endocrinology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Ying Hu
- Medical School of Yangtze University, Jingzhou, China
| | - Jiao Yang
- Medical School of Yangtze University, Jingzhou, China
| | | | - Xiaochun Peng
- Medical School of Yangtze University, Jingzhou, China,
| |
Collapse
|
32
|
Chait A, den Hartigh LJ. Adipose Tissue Distribution, Inflammation and Its Metabolic Consequences, Including Diabetes and Cardiovascular Disease. Front Cardiovasc Med 2020; 7:22. [PMID: 32158768 PMCID: PMC7052117 DOI: 10.3389/fcvm.2020.00022] [Citation(s) in RCA: 633] [Impact Index Per Article: 158.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/10/2020] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue plays essential roles in maintaining lipid and glucose homeostasis. To date several types of adipose tissue have been identified, namely white, brown, and beige, that reside in various specific anatomical locations throughout the body. The cellular composition, secretome, and location of these adipose depots define their function in health and metabolic disease. In obesity, adipose tissue becomes dysfunctional, promoting a pro-inflammatory, hyperlipidemic and insulin resistant environment that contributes to type 2 diabetes mellitus (T2DM). Concurrently, similar features that result from adipose tissue dysfunction also promote cardiovascular disease (CVD) by mechanisms that can be augmented by T2DM. The mechanisms by which dysfunctional adipose tissue simultaneously promote T2DM and CVD, focusing on adipose tissue depot-specific adipokines, inflammatory profiles, and metabolism, will be the focus of this review. The impact that various T2DM and CVD treatment strategies have on adipose tissue function and body weight also will be discussed.
Collapse
Affiliation(s)
- Alan Chait
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Laura J den Hartigh
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
33
|
Endogenous FGF21-signaling controls paradoxical obesity resistance of UCP1-deficient mice. Nat Commun 2020; 11:624. [PMID: 32005798 PMCID: PMC6994690 DOI: 10.1038/s41467-019-14069-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 12/10/2019] [Indexed: 01/01/2023] Open
Abstract
Uncoupling protein 1 (UCP1) executes thermogenesis in brown adipose tissue, which is a major focus of human obesity research. Although the UCP1-knockout (UCP1 KO) mouse represents the most frequently applied animal model to judge the anti-obesity effects of UCP1, the assessment is confounded by unknown anti-obesity factors causing paradoxical obesity resistance below thermoneutral temperatures. Here we identify the enigmatic factor as endogenous FGF21, which is primarily mediating obesity resistance. The generation of UCP1/FGF21 double-knockout mice (dKO) fully reverses obesity resistance. Within mild differences in energy metabolism, urine metabolomics uncover increased secretion of acyl-carnitines in UCP1 KOs, suggesting metabolic reprogramming. Strikingly, transcriptomics of metabolically important organs reveal enhanced lipid and oxidative metabolism in specifically white adipose tissue that is fully reversed in dKO mice. Collectively, this study characterizes the effects of endogenous FGF21 that acts as master regulator to protect from diet-induced obesity in the absence of UCP1. Brown adipose thermogenesis increases energy expenditure and relies on uncoupling protein 1 (UCP1), however, UCP1 knock-out mice show resistance to diet-induced obesity at room temperature. Here, the authors show that this resistance relies on FGF21-signaling, inducing the browning of white adipose tissue.
Collapse
|
34
|
Non-canonical mTORC2 Signaling Regulates Brown Adipocyte Lipid Catabolism through SIRT6-FoxO1. Mol Cell 2020; 75:807-822.e8. [PMID: 31442424 DOI: 10.1016/j.molcel.2019.07.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/30/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022]
Abstract
mTORC2 controls glucose and lipid metabolism, but the mechanisms are unclear. Here, we show that conditionally deleting the essential mTORC2 subunit Rictor in murine brown adipocytes inhibits de novo lipid synthesis, promotes lipid catabolism and thermogenesis, and protects against diet-induced obesity and hepatic steatosis. AKT kinases are the canonical mTORC2 substrates; however, deleting Rictor in brown adipocytes appears to drive lipid catabolism by promoting FoxO1 deacetylation independently of AKT, and in a pathway distinct from its positive role in anabolic lipid synthesis. This facilitates FoxO1 nuclear retention, enhances lipid uptake and lipolysis, and potentiates UCP1 expression. We provide evidence that SIRT6 is the FoxO1 deacetylase suppressed by mTORC2 and show an endogenous interaction between SIRT6 and mTORC2 in both mouse and human cells. Our findings suggest a new paradigm of mTORC2 function filling an important gap in our understanding of this more mysterious mTOR complex.
Collapse
|
35
|
Scheele C, Wolfrum C. Brown Adipose Crosstalk in Tissue Plasticity and Human Metabolism. Endocr Rev 2020; 41:bnz007. [PMID: 31638161 PMCID: PMC7006230 DOI: 10.1210/endrev/bnz007] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 10/15/2019] [Indexed: 12/20/2022]
Abstract
Infants rely on brown adipose tissue (BAT) as a primary source of thermogenesis. In some adult humans, residuals of brown adipose tissue are adjacent to the central nervous system and acute activation increases metabolic rate. Brown adipose tissue (BAT) recruitment occurs during cold acclimation and includes secretion of factors, known as batokines, which target several different cell types within BAT, and promote adipogenesis, angiogenesis, immune cell interactions, and neurite outgrowth. All these processes seem to act in concert to promote an adapted BAT. Recent studies have also provided exciting data on whole body metabolic regulation with a broad spectrum of mechanisms involving BAT crosstalk with liver, skeletal muscle, and gut as well as the central nervous system. These widespread interactions might reflect the property of BAT of switching between an active thermogenic state where energy is highly consumed and drained from the circulation, and the passive thermoneutral state, where energy consumption is turned off. (Endocrine Reviews 41: XXX - XXX, 2020).
Collapse
Affiliation(s)
- Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, Denmark
| | - Christian Wolfrum
- Institute of Food, Nutrition, and Health, ETH Zürich, Schorenstrasse, Schwerzenbach, Switzerland
| |
Collapse
|
36
|
Leiva M, Matesanz N, Pulgarín-Alfaro M, Nikolic I, Sabio G. Uncovering the Role of p38 Family Members in Adipose Tissue Physiology. Front Endocrinol (Lausanne) 2020; 11:572089. [PMID: 33424765 PMCID: PMC7786386 DOI: 10.3389/fendo.2020.572089] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
The complex functions of adipose tissue have been a focus of research interest over the past twenty years. Adipose tissue is not only the main energy storage depot, but also one of the largest endocrine organs in the body and carries out crucial metabolic functions. Moreover, brown and beige adipose depots are major sites of energy expenditure through the activation of adaptive, non-shivering thermogenesis. In recent years, numerous signaling molecules and pathways have emerged as critical regulators of adipose tissue, in both homeostasis and obesity-related disease. Among the best characterized are members of the p38 kinase family. The activity of these kinases has emerged as a key contributor to the biology of the white and brown adipose tissues, and their modulation could provide new therapeutic approaches against obesity. Here, we give an overview of the roles of the distinct p38 family members in adipose tissue, focusing on their actions in adipogenesis, thermogenic activity, and secretory function.
Collapse
|
37
|
Lelis DDF, Freitas DFD, Machado AS, Crespo TS, Santos SHS. Angiotensin-(1-7), Adipokines and Inflammation. Metabolism 2019; 95:36-45. [PMID: 30905634 DOI: 10.1016/j.metabol.2019.03.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/11/2019] [Accepted: 03/20/2019] [Indexed: 12/12/2022]
Abstract
Nowadays the adipose tissue is recognized as one of the most critical endocrine organs releasing many adipokines that regulate metabolism, inflammation and body homeostasis. There are several described adipokines, including the renin-angiotensin system (RAS) components that are especially activated in some diseases with increased production of angiotensin II and several pro-inflammatory hormones. On the other hand, RAS also expresses angiotensin-(1-7), which is now recognized as the main peptide on counteracting Ang II effects. New studies have shown that increased activation of ACE2/Ang-(1-7)/MasR arm can revert and prevent local and systemic dysfunctions improving lipid profile and insulin resistance by modulating insulin actions, and reducing inflammation. In this context, the present review shows the interaction and relevance of Ang-(1-7) effects on regulating adipokines, and as one adipokine itself, modulating body homeostasis, with emphasis on its anti-inflammatory properties, especially in the context of metabolic disorders with focus on obesity and type 2 diabetes mellitus pandemic.
Collapse
Affiliation(s)
- Deborah de Farias Lelis
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Daniela Fernanda de Freitas
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Amanda Souto Machado
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Thaísa Soares Crespo
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Institute of Agricultural Sciences, Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil; Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil.
| |
Collapse
|
38
|
Loss of TLE3 promotes the mitochondrial program in beige adipocytes and improves glucose metabolism. Genes Dev 2019; 33:747-762. [PMID: 31123067 PMCID: PMC6601513 DOI: 10.1101/gad.321059.118] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 04/11/2019] [Indexed: 11/25/2022]
Abstract
In this study, Pearson et al. investigated the transcriptional mechanisms that promote remodeling in adipose tissue during the cold. Their findings demonstrate that transcriptional coregulator TLE3 regulates thermogenic gene expression in beige adipocytes through inhibition of EBF2 transcriptional activity. Prolonged cold exposure stimulates the recruitment of beige adipocytes within white adipose tissue. Beige adipocytes depend on mitochondrial oxidative phosphorylation to drive thermogenesis. The transcriptional mechanisms that promote remodeling in adipose tissue during the cold are not well understood. Here we demonstrate that the transcriptional coregulator transducin-like enhancer of split 3 (TLE3) inhibits mitochondrial gene expression in beige adipocytes. Conditional deletion of TLE3 in adipocytes promotes mitochondrial oxidative metabolism and increases energy expenditure, thereby improving glucose control. Using chromatin immunoprecipitation and deep sequencing, we found that TLE3 occupies distal enhancers in proximity to nuclear-encoded mitochondrial genes and that many of these binding sites are also enriched for early B-cell factor (EBF) transcription factors. TLE3 interacts with EBF2 and blocks its ability to promote the thermogenic transcriptional program. Collectively, these studies demonstrate that TLE3 regulates thermogenic gene expression in beige adipocytes through inhibition of EBF2 transcriptional activity. Inhibition of TLE3 may provide a novel therapeutic approach for obesity and diabetes.
Collapse
|
39
|
Interscapular and Perivascular Brown Adipose Tissue Respond Differently to a Short-Term High-Fat Diet. Nutrients 2019; 11:nu11051065. [PMID: 31086124 PMCID: PMC6566556 DOI: 10.3390/nu11051065] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
Brown adipose tissue (BAT) function may depend on its anatomical location and developmental origin. Interscapular BAT (iBAT) regulates acute macronutrient metabolism, whilst perivascular BAT (PVAT) regulates vascular function. Although phenotypically similar, whether these depots respond differently to acute nutrient excess is unclear. Given their distinct anatomical locations and developmental origins and we hypothesised that iBAT and PVAT would respond differently to brief period of nutrient excess. Sprague-Dawley rats aged 12 weeks (n=12) were fed either a standard (10% fat, n=6) or high fat diet (HFD: 45% fat, n=6) for 72h and housed at thermoneutrality. Following an assessment of whole body physiology, fat was collected from both depots for analysis of gene expression and the proteome. HFD consumption for 72h induced rapid weight gain (c. 2.6%) and reduced serum non-esterified fatty acids (NEFA) with no change in either total adipose or depot mass. In iBAT, an upregulation of genes involved in insulin signalling and lipid metabolism was accompanied by enrichment of lipid-related processes and functions, plus glucagon and peroxisome proliferator-activated receptor (PPAR) signalling pathways. In PVAT, HFD induced a pronounced down-regulation of multiple metabolic pathways which was accompanied with increased abundance of proteins involved in apoptosis (e.g., Hdgf and Ywaq) and toll-like receptor signalling (Ube2n). There was also an enrichment of DNA-related processes and functions (e.g., nucleosome assembly and histone exchange) and RNA degradation and cell adhesion pathways. In conclusion, we show that iBAT and PVAT elicit divergent responses to short-term nutrient excess highlighting early adaptations in these depots before changes in fat mass.
Collapse
|
40
|
Fukui K, Shirai M, Ninuma T, Kato Y. Anti-Obesity Effects of Tocotrienols and Bran in High-Fat Diet-Treated Mice. Nutrients 2019; 11:nu11040830. [PMID: 31013725 PMCID: PMC6521046 DOI: 10.3390/nu11040830] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/05/2019] [Accepted: 04/08/2019] [Indexed: 01/17/2023] Open
Abstract
Obesity is a serious public health issue in developed countries, and is known to increase the risk of several diseases such as diabetes, cardiovascular events and arteriosclerosis. These phenomena are closely correlated with oxidative damage. Recently, several lines of evidence have demonstrated that neurodegenerative diseases such as Alzheimer’s and Parkinson’s are also related to oxidative damage. To clarify the relationship between obesity and oxidative brain injury, we investigated brain antioxidant networks in high-fat (HF) diet-treated mice in the presence or absence of tocotrienols (T3s) and bran. Co-treatment with T3s and bran significantly inhibited bodyweight gain in HF diet-treated mice. Serum and cortex T3 levels, and brain antioxidant enzyme activities and protein expressions did not differ among the groups except for SOD protein expression in the cerebellum. Brain p-mTOR and p-Akt protein expressions, which are related to autophagy, did not differ among the groups. These results indicate that treatment with T3s for eight weeks had showed an anti-obesity effect in HF diet-treated mice. However, significant alterations in T3 levels were not observed in the serum and brain of mice.
Collapse
Affiliation(s)
- Koji Fukui
- Molecular Cell Biology Laboratory, Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan.
| | - Masashi Shirai
- Molecular Cell Biology Laboratory, Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan.
| | - Takeyuki Ninuma
- Molecular Cell Biology Laboratory, Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan.
| | - Yugo Kato
- Molecular Cell Biology Laboratory, Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan.
| |
Collapse
|
41
|
Jespersen NZ, Feizi A, Andersen ES, Heywood S, Hattel HB, Daugaard S, Peijs L, Bagi P, Feldt-Rasmussen B, Schultz HS, Hansen NS, Krogh-Madsen R, Pedersen BK, Petrovic N, Nielsen S, Scheele C. Heterogeneity in the perirenal region of humans suggests presence of dormant brown adipose tissue that contains brown fat precursor cells. Mol Metab 2019; 24:30-43. [PMID: 31079959 PMCID: PMC6531810 DOI: 10.1016/j.molmet.2019.03.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/02/2019] [Accepted: 03/11/2019] [Indexed: 12/31/2022] Open
Abstract
Objective Increasing the amounts of functionally competent brown adipose tissue (BAT) in adult humans has the potential to restore dysfunctional metabolism and counteract obesity. In this study, we aimed to characterize the human perirenal fat depot, and we hypothesized that there would be regional, within-depot differences in the adipose signature depending on local sympathetic activity. Methods We characterized fat specimens from four different perirenal regions of adult kidney donors, through a combination of qPCR mapping, immunohistochemical staining, RNA-sequencing, and pre-adipocyte isolation. Candidate gene signatures, separated by adipocyte morphology, were recapitulated in a murine model of unilocular brown fat induced by thermoneutrality and high fat diet. Results We identified widespread amounts of dormant brown adipose tissue throughout the perirenal depot, which was contrasted by multilocular BAT, primarily found near the adrenal gland. Dormant BAT was characterized by a unilocular morphology and a distinct gene expression profile, which partly overlapped with that of subcutaneous white adipose tissue (WAT). Brown fat precursor cells, which differentiated into functional brown adipocytes were present in the entire perirenal fat depot, regardless of state. We identified SPARC as a candidate adipokine contributing to a dormant BAT state, and CLSTN3 as a novel marker for multilocular BAT. Conclusions We propose that perirenal adipose tissue in adult humans consists mainly of dormant BAT and provide a data set for future research on factors which can reactivate dormant BAT into active BAT, a potential strategy for combatting obesity and metabolic disease. Dormant brown adipose tissue (BAT) dominates the perirenal adipose depot of adult humans. Multilocular BAT accumulates adjacent to local sympathetic sources. Dormant BAT displays a transcriptomic signature distinct from multilocular BAT and white adipose tissue. Brown fat precursor cells are present in human dormant perirenal BAT. SPARC is associated with a dormant BAT phenotype.
Collapse
Affiliation(s)
- Naja Z Jespersen
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, 2100, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Danish PhD School of Molecular Metabolism, Odense, Denmark
| | - Amir Feizi
- Novo Nordisk Research Center Oxford, Denmark
| | - Eline S Andersen
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, 2100, Denmark
| | - Sarah Heywood
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, 2100, Denmark
| | - Helle B Hattel
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, 2100, Denmark
| | | | - Lone Peijs
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, 2100, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Per Bagi
- Department of Urology, Rigshospitalet, Denmark
| | | | | | - Ninna S Hansen
- Danish PhD School of Molecular Metabolism, Odense, Denmark; Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, Denmark
| | - Rikke Krogh-Madsen
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, 2100, Denmark
| | - Bente K Pedersen
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, 2100, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Natasa Petrovic
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, 106 91, Stockholm, Sweden
| | - Søren Nielsen
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, 2100, Denmark
| | - Camilla Scheele
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, 2100, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
42
|
Ro SH, Jang Y, Bae J, Kim IM, Schaecher C, Shomo ZD. Autophagy in Adipocyte Browning: Emerging Drug Target for Intervention in Obesity. Front Physiol 2019; 10:22. [PMID: 30745879 PMCID: PMC6360992 DOI: 10.3389/fphys.2019.00022] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 01/10/2019] [Indexed: 12/27/2022] Open
Abstract
Autophagy, lipophagy, and mitophagy are considered to be the major recycling processes for protein aggregates, excess fat, and damaged mitochondria in adipose tissues in response to nutrient status-associated stress, oxidative stress, and genotoxic stress in the human body. Obesity with increased body weight is often associated with white adipose tissue (WAT) hypertrophy and hyperplasia and/or beige/brown adipose tissue atrophy and aplasia, which significantly contribute to the imbalance in lipid metabolism, adipocytokine secretion, free fatty acid release, and mitochondria function. In recent studies, hyperactive autophagy in WAT was observed in obese and diabetic patients, and inhibition of adipose autophagy through targeted deletion of autophagy genes in mice improved anti-obesity phenotypes. In addition, active mitochondria clearance through activation of autophagy was required for beige/brown fat whitening – that is, conversion to white fat. However, inhibition of autophagy seemed detrimental in hypermetabolic conditions such as hepatic steatosis, atherosclerosis, thermal injury, sepsis, and cachexia through an increase in free fatty acid and glycerol release from WAT. The emerging concept of white fat browning–conversion to beige/brown fat–has been controversial in its anti-obesity effect through facilitation of weight loss and improving metabolic health. Thus, proper regulation of autophagy activity fit to an individual metabolic profile is necessary to ensure balance in adipose tissue metabolism and function, and to further prevent metabolic disorders such as obesity and diabetes. In this review, we summarize the effect of autophagy in adipose tissue browning in the context of obesity prevention and its potential as a promising target for the development of anti-obesity drugs.
Collapse
Affiliation(s)
- Seung-Hyun Ro
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Yura Jang
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jiyoung Bae
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States.,Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Isaac M Kim
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Cameron Schaecher
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States.,College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Zachery D Shomo
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
43
|
Tatenaka Y, Kato H, Ishiyama M, Sasamoto K, Shiga M, Nishitoh H, Ueno Y. Monitoring Lipid Droplet Dynamics in Living Cells by Using Fluorescent Probes. Biochemistry 2019; 58:499-503. [PMID: 30628446 DOI: 10.1021/acs.biochem.8b01071] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We have developed three types of lipid droplet (LD)-specific fluorescent probes for live-cell imaging, Lipi-Blue, Lipi-Green, and Lipi-Red, which exhibit fluorescence upon being incorporated into LDs both of living and of fixed cells. These Lipi-probes are LD-specific probes that contain a pyrene or perylene group as a fluorescent scaffold and can be used to observe dynamics of LD in live cells and also interrelations with other organelles by simultaneous staining with multiple organelle-specific probes. Additionally, Lipi-Blue and Lipi-Green allow monitoring LDs in live cells even for 48 h after the staining. Here we show that newly formed LDs and previously existed LDs can be separately monitored in a single cell by using these probes and that intercellular transfer of whole LDs is observed in KB cells, but not in HepG2 cells under the same culturing condition. These findings indicate that newly developed LD-specific probes are useful to analyze the dynamics of LDs in live cells.
Collapse
Affiliation(s)
- Yuki Tatenaka
- Dojindo Laboratories , Tabaru 2025-5 , Mashiki-machi , Kumamoto 861-2202 , Japan
| | - Hironori Kato
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Sciences , University of Miyazaki , 5200 Kihara Kiyotake , Miyazaki 889-1692 , Japan
| | - Munetaka Ishiyama
- Dojindo Laboratories , Tabaru 2025-5 , Mashiki-machi , Kumamoto 861-2202 , Japan
| | - Kazumi Sasamoto
- Dojindo Laboratories , Tabaru 2025-5 , Mashiki-machi , Kumamoto 861-2202 , Japan
| | - Masanobu Shiga
- Dojindo Laboratories , Tabaru 2025-5 , Mashiki-machi , Kumamoto 861-2202 , Japan
| | - Hideki Nishitoh
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Sciences , University of Miyazaki , 5200 Kihara Kiyotake , Miyazaki 889-1692 , Japan
| | - Yuichiro Ueno
- Dojindo Laboratories , Tabaru 2025-5 , Mashiki-machi , Kumamoto 861-2202 , Japan
| |
Collapse
|
44
|
Di Gregorio I, Busiello RA, Burgos Aceves MA, Lepretti M, Paolella G, Lionetti L. Environmental Pollutants Effect on Brown Adipose Tissue. Front Physiol 2019; 9:1891. [PMID: 30687113 PMCID: PMC6333681 DOI: 10.3389/fphys.2018.01891] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 12/14/2018] [Indexed: 12/19/2022] Open
Abstract
Brown adipose tissue (BAT) with its thermogenic function due to the presence of the mitochondrial uncoupling protein 1 (UCP1), has been positively associated with improved resistance to obesity and metabolic diseases. During recent years, the potential influence of environmental pollutants on energetic homoeostasis and obesity development has drawn increased attention. The purpose of this review is to discuss how regulation of BAT function could be involved in the environmental pollutant effect on body energy metabolism. We mainly focused in reviewing studies on animal models, which provide a better insight into the cellular mechanisms involved in this effect on body energy metabolism. The current literature supports the hypothesis that some environmental pollutants, acting as endocrine disruptors (EDCs), such as dichlorodiphenyltrichoroethane (DDT) and its metabolite dichlorodiphenylethylene (DDE) as well as some, traffic pollutants, are associated with increased obesity risk, whereas some other chemicals, such as perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA), had a reverse association with obesity. Noteworthy, the EDCs associated with obesity and metabolic disorders impaired BAT mass and function. Perinatal exposure to DDT impaired BAT thermogenesis and substrate utilization, increasing susceptibility to metabolic syndrome. Ambient particulate air pollutions induced insulin resistance associated with BAT mitochondrial dysfunction. On the other hand, the environmental pollutants (PFOS/PFOA) elicited a reduction in body weight and adipose mass associated with upregulation of UCP1 and increased oxidative capacity in brown-fat mitochondria. Further research is needed to better understand the physiological role of BAT in response to exposure to both obesogenic and anti-obesogenic pollutants and to confirm the same role in humans.
Collapse
Affiliation(s)
| | | | | | | | | | - Lillà Lionetti
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, Fisciano, Italy
| |
Collapse
|
45
|
McGeoch PD. Can Vestibular Stimulation be Used to Treat Obesity? Bioessays 2019; 41:e1800197. [DOI: 10.1002/bies.201800197] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/04/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Paul D. McGeoch
- Center for Brain and Cognition; UC San Diego, Mandler Hall; 9500 Gilman Drive La Jolla CA 92093 US
| |
Collapse
|
46
|
Silvester AJ, Aseer KR, Jang HJ, Ryu R, Kwon EY, Park JG, Cho KH, Chaudhari HN, Choi MS, Suh PG, Yun JW. Loss of DJ-1 promotes browning of white adipose tissue in diet-induced obese mice. J Nutr Biochem 2018; 61:56-67. [PMID: 30189364 DOI: 10.1016/j.jnutbio.2018.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/21/2018] [Indexed: 11/26/2022]
Abstract
The seminal discovery of browning of white adipose tissue (WAT) holds great promise for the treatment of obesity and metabolic syndrome. DJ-1 is evolutionarily conserved across species, and mutations in DJ-1 have been identified in Parkinson's disease. Higher levels of DJ-1 are associated with obesity, but the underlying mechanism is less understood. Here, we report the previously unappreciated role of DJ-1 in white adipocyte biology in mature models of obesity. We used DJ-1 knockout (KO) mouse models and wild-type littermates maintained on a normal diet or high-fat diet as well as in vitro cell models to show the direct effects of DJ-1 depletion on adipocyte phenotype, thermogenic capacity, fat metabolism, and microenvironment profile. Global DJ-1 KO mice show increased sympathetic input to WAT and β3-adrenergic receptor intracellular signaling, leading to a previously unrecognized compensatory mechanism through browning of WAT with associated characteristics, including high mitochondrial contents, reduced lipid accumulation, adequate vascularization and attenuated autophagy. DJ-1 KO mice had normal body weight, energy balance, and adiposity, which were associated with protective effects on healthy WAT expansion by hyperplasia. Our findings revealed that browning of inguinal WAT occurred in DJ-1 KO mice that do not show increased predisposition to obesity and suggest that such potential mechanism may overcome the adverse metabolic consequences of obesity independent of an effect on body weight. Here, we provide the first direct evidence that targeting DJ-1 in adipocyte metabolic health may offer a unique therapeutic strategy for the treatment of obesity.
Collapse
Affiliation(s)
| | - Kanikkai Raja Aseer
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea
| | - Hyun-Jun Jang
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Ri Ryu
- Department of Food Science and Nutrition, Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Eun-Young Kwon
- Department of Food Science and Nutrition, Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jae Gyu Park
- Advanced Bio Convergence Center, Pohang Technopark Foundation, Pohang 37668, Republic of Korea
| | - Kiu-Hyung Cho
- Gyeongbuk Institute for Bioindustry, Andong 31984, Republic of Korea
| | - Harmesh N Chaudhari
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea
| | - Myung-Sook Choi
- Department of Food Science and Nutrition, Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Pann-Ghill Suh
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Jong Won Yun
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| |
Collapse
|
47
|
Carpentier AC, Blondin DP, Virtanen KA, Richard D, Haman F, Turcotte ÉE. Brown Adipose Tissue Energy Metabolism in Humans. Front Endocrinol (Lausanne) 2018; 9:447. [PMID: 30131768 PMCID: PMC6090055 DOI: 10.3389/fendo.2018.00447] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/20/2018] [Indexed: 12/16/2022] Open
Abstract
The demonstration of metabolically active brown adipose tissue (BAT) in humans primarily using positron emission tomography coupled to computed tomography (PET/CT) with the glucose tracer 18-fluorodeoxyglucose (18FDG) has renewed the interest of the scientific and medical community in the possible role of BAT as a target for the prevention and treatment of obesity and type 2 diabetes (T2D). Here, we offer a comprehensive review of BAT energy metabolism in humans. Considerable advances in methods to measure BAT energy metabolism, including nonesterified fatty acids (NEFA), chylomicron-triglycerides (TG), oxygen, Krebs cycle rate, and intracellular TG have led to very good quantification of energy substrate metabolism per volume of active BAT in vivo. These studies have also shown that intracellular TG are likely the primary energy source of BAT upon activation by cold. Current estimates of BAT's contribution to energy expenditure range at the lower end of what would be potentially clinically relevant if chronically sustained. Yet, 18FDG PET/CT remains the gold-standard defining method to quantify total BAT volume of activity, used to calculate BAT's total energy expenditure. Unfortunately, BAT glucose metabolism better reflects BAT's insulin sensitivity and blood flow. It is now clear that most glucose taken up by BAT does not fuel mitochondrial oxidative metabolism and that BAT glucose uptake can therefore be disconnected from thermogenesis. Furthermore, BAT thermogenesis is efficiently recruited upon repeated cold exposure, doubling to tripling its total oxidative capacity, with reciprocal reduction of muscle thermogenesis. Recent data suggest that total BAT volume may be much larger than the typically observed 50-150 ml with 18FDG PET/CT. Therefore, the current estimates of total BAT thermogenesis, largely relying on total BAT volume using 18FDG PET/CT, may underestimate the true contribution of BAT to total energy expenditure. Quantification of the contribution of BAT to energy expenditure begs for the development of more integrated whole body in vivo methods.
Collapse
Affiliation(s)
- André C. Carpentier
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Kirsi A. Virtanen
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland (UEF), Kuopio, Finland
| | - Denis Richard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, QC, Canada
| | - François Haman
- Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Éric E. Turcotte
- Department of Nuclear Medicine and Radiobiology, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
48
|
Pyruvate dehydrogenase complex plays a central role in brown adipocyte energy expenditure and fuel utilization during short-term beta-adrenergic activation. Sci Rep 2018; 8:9562. [PMID: 29934543 PMCID: PMC6015083 DOI: 10.1038/s41598-018-27875-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/12/2018] [Indexed: 12/27/2022] Open
Abstract
Activation of brown adipose tissue (BAT) contributes to total body energy expenditure through energy dissipation as heat. Activated BAT increases the clearance of lipids and glucose from the circulation, but how BAT accommodates large influx of multiple substrates is not well defined. The purpose of this work was to assess the metabolic fluxes in brown adipocytes during β3-adrenergic receptor (β3-AR) activation.T37i murine preadipocytes were differentiated into brown adipocytes and we used Seahorse respirometry employing a set of specific substrate inhibitors in the presence or absence of β3-AR agonist CL316,243. The main substrate used by these brown adipocytes were fatty acids, which were oxidized equally during activation as well as during resting condition. [U-13C]-glucose tracer-based metabolomics revealed that the flux through the TCA cycle was enhanced and regulated by pyruvate dehydrogenase (PDH) activity. Based on 13C-tracer incorporation in lipids, it appeared that most glucose was oxidized via TCA cycle activity, while some was utilized for glycerol-3-phosphate synthesis to replenish the triglyceride pool. Collectively, we show that while fatty acids are the main substrates for oxidation, glucose is also oxidized to meet the increased energy demand during short term β3-AR activation. PDH plays an important role in directing glucose carbons towards oxidation.
Collapse
|
49
|
Effect of long-term nitrite administration on browning of white adipose tissue in type 2 diabetic rats: A stereological study. Life Sci 2018; 207:219-226. [PMID: 29898382 DOI: 10.1016/j.lfs.2018.06.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/27/2018] [Accepted: 06/09/2018] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Nitric oxide (NO) deficiency is associated with obesity and type 2 diabetes. Nitrite, a NO donor, is considered as a new therapeutic agent in diabetes. This study aims at determining effects of long-term nitrite administration on browning of white adipose tissue (WAT) in type 2 diabetic rats. METHODS Male rats were divided into 4 groups: Control, control + nitrite, diabetes, and diabetes + nitrite. Sodium nitrite (50 mg/L in drinking water) was administered for 3 months. Body weight was measured weekly. Fasting serum levels of glucose and nitric oxide metabolites (NOx) were measured monthly. Histological evaluations and measurement of cyclic guanosine monophosphate (cGMP) and NOx levels in adipose tissue were done at the end of the study. RESULTS Nitrite decreased serum glucose concentration and body weight gain in diabetic rats by 27.6% and 37.9%, respectively. In diabetic rats, nitrite increased NOx and cGMP levels in inguinal WAT by 95.7% and 33.1%, respectively. Numerical density in WAT of nitrite-treated diabetic rats was higher than in diabetic ones (995 ± 83 vs. 2513 ± 256 cell/mm3, P < 0.001); in addition, total surface area (4.84 ± 0.32 vs. 44.26 ± 9.7, mm2, P < 0.001) and volume of inguinal beige adipose tissue (7.2 ± 0.49 vs. 66.4 ± 14.51 mm3, <0.001) were higher in nitrite-treated diabetic rats compared to diabetic ones. CONCLUSIONS Favorable effects of long-term nitrite administration in obese type 2 diabetic rats is, at least in part, due to browning of WAT and also associated with increased NOx and cGMP level in adipose tissue. These findings may have potential applications for management of diabesity.
Collapse
|
50
|
Abstract
Numerous studies have shown that feeding rodents n-3 polyunsaturated fatty acids attenuates adiposity. Moreover, meta-analyses of human dietary intervention studies indicate that fish oil (eicosapentaenoic and docosahexaenoic acid) supplementation might reduce waist circumference. A recent line of research suggests that browning of white adipose depots and activation of uncoupled respiration in brown fat contributes to these effects. This mini-review summarizes the observations in rodents, highlights several mechanisms that might explain these observations and discusses the translational potential. Given the available in vivo evidence and the ability of human adipocytes to aquire a beige phenotype in response to eicosapentaenoic acid incubation, future studies should test the hypothesis that fish oil activates thermogenic brown and beige adipose tissue in humans.
Collapse
Affiliation(s)
- Jens Lund
- Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Lesli Hingstrup Larsen
- Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Lotte Lauritzen
- Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|