1
|
Chilamakuri SN, N M, Thalla M, Velayutham R, Lee Y, Cho SM, Jung H, Natesan S. Role of Microneedles for Improved Treatment of Obesity: Progress and Challenges. Mol Pharm 2025; 22:2350-2371. [PMID: 40167034 DOI: 10.1021/acs.molpharmaceut.4c01115] [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: 04/02/2025]
Abstract
Obesity is a global metabolic health epidemic characterized by excessive lipid and fat accumulation, leading to severe conditions such as diabetes, cancer, and cardiovascular disease. Immediate attention and management of obesity-related health risks are most warranted. The imbalance between fat absorption, metabolic rate, and environmental and genetic factors is responsible for obesity. Treatment typically involves lifestyle modifications, pharmacotherapy, and surgery. While lifestyle changes are crucial, effective treatment often necessitates medication as a preferred adjunct strategy. However, medications commonly used, such as oral pharmacotherapy, often show side effects due to systemic exposure and, thus, may not effectively target the intended areas, leading to drug loss. On the other hand, transdermal administration of drugs with microneedle (MN)-based technologies, a painless drug delivery approach with patient compliance, is gaining interest as an alternative obesity treatment, as it directly targets adipose tissue via local delivery, minimizing system exposure and dose reduction. This Review addresses the pathophysiology of obesity, current treatment strategies, challenges in the treatment of obesity using conventional formulations, the importance of the use of nano-based medications through transdermal delivery, and the use of MNs as a promising platform for the effective delivery of nanoparticle-based anti-obesity medications. The potential of combining MNs with stimuli-responsive and non-responsive adjuvant therapies to enhance treatment efficacy and patient outcomes is explored. In addition, the limitations and future perspectives related to the use of MNs for obesity are addressed to highlight the transformative potential of this technology for obesity management. MNs hold promise in precisely delivering anti-obesity drugs while requiring lower dosages and minimizing side effects compared to conventional oral or injectable therapies and ultimately improving the quality of life for individuals struggling with obesity and its associated comorbidities.
Collapse
Affiliation(s)
- Sudarshan Naidu Chilamakuri
- Department of Pharmaceutics, Advance Formulation Laboratory, National Institute of Pharmaceutical Education and Research, Kolkata, 700054, West Bengal, India
| | - Manasa N
- Department of Pharmaceutics, Advance Formulation Laboratory, National Institute of Pharmaceutical Education and Research, Kolkata, 700054, West Bengal, India
| | - Maharshi Thalla
- Department of Pharmaceutical Sciences, Texas A&M University, Kingsville, Texas 78363, United States
| | - Ravichandiran Velayutham
- Department of Pharmaceutics, Advance Formulation Laboratory, National Institute of Pharmaceutical Education and Research, Kolkata, 700054, West Bengal, India
| | - Youjin Lee
- Department of Integrative Biotechnology, Yonsei University, 85 Songdogwahak-ro, Incheon 21983, Republic of Korea
| | - Sung Min Cho
- Department of Integrative Biotechnology, Yonsei University, 85 Songdogwahak-ro, Incheon 21983, Republic of Korea
| | - Hyungil Jung
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seoul 08389, Republic of Korea
- Department of Integrative Biotechnology, Yonsei University, 85 Songdogwahak-ro, Incheon 21983, Republic of Korea
| | - Subramanian Natesan
- Department of Pharmaceutics, Advance Formulation Laboratory, National Institute of Pharmaceutical Education and Research, Kolkata, 700054, West Bengal, India
| |
Collapse
|
2
|
Futatsugi M, Miyazaki A, Kanai Y, Kondo N, Temma T. Fluorine-18-Labeled Positron Emission Tomography Probe Targeting Activated p38α: Design, Synthesis, and In Vivo Evaluation in Rodents. Pharmaceuticals (Basel) 2025; 18:600. [PMID: 40284035 PMCID: PMC12030359 DOI: 10.3390/ph18040600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: The kinase p38α, a member of the mitogen-activated protein kinase (MAPK) family, is activated by external stimuli and plays a crucial role in inflammation, tumor growth, and metabolic disorders. In particular, p38α is involved in thermogenesis and the metabolism of glucose in brown adipose tissue (BAT), and it contributes to the suppression of obesity and diabetes. The noninvasive imaging of activated p38α could help elucidate diverse pathological processes, including metabolic and inflammatory conditions. This study aimed to develop and evaluate a novel fluorine-18-labeled positron emission tomography (PET) probe for imaging activated p38α in vivo. Methods: We designed 6-(4-[18F]fluoro-2-fluorophenoxy)-8-methyl-2-(tetrahydro-2H-pyran-4-ylamino)-pyrido[2,3-d]pyrimidin-7(8H)-one ([18F]R1487) by replacing a fluorine atom in R1487, which is a highly selective p38α inhibitor, with 18F. A tributylstannyl precursor was reacted with [18F]KF in the presence of a copper catalyst to synthesize [18F]R1487. Biodistribution studies and PET/computed tomography (CT) were performed on normal mice to evaluate the in vivo potential of [18F]R1487. Results: [18F]R1487 was obtained with a decay-corrected radiochemical conversion of 30.6 ± 5.6% and a decay-corrected radiochemical yield of 6.9 ± 3.6% with a radiochemical purity of >99% after reversed-phase high-performance liquid chromatography purification. The biodistribution study demonstrated high and rapid radioactivity accumulation in BAT (16.3 ± 2.7 %ID/g at 5 min post-injection), with a consistently high BAT-to-blood ratio (>5 over 2 h post-injection). PET/CT imaging successfully visualized BAT with high contrast. Conclusions: These results suggest that [18F]R1487 is a promising PET probe for imaging activated p38α in vivo, which has potential applications for pathophysiological conditions such as inflammation, cancer, and metabolic disorders.
Collapse
Affiliation(s)
- Mikiya Futatsugi
- Department of Biofunctional Analysis, Graduate School of Pharmaceutical Sciences, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki 569-1094, Osaka, Japan; (M.F.); (A.M.); (N.K.)
| | - Anna Miyazaki
- Department of Biofunctional Analysis, Graduate School of Pharmaceutical Sciences, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki 569-1094, Osaka, Japan; (M.F.); (A.M.); (N.K.)
| | - Yasukazu Kanai
- Kansai BNCT Medical Center, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan;
| | - Naoya Kondo
- Department of Biofunctional Analysis, Graduate School of Pharmaceutical Sciences, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki 569-1094, Osaka, Japan; (M.F.); (A.M.); (N.K.)
- Division of Fundamental Technology Development, Near InfraRed Photo-ImmunoTherapy Research Institute, Kansai Medical University, 2-5-1 Shin-machi, Hirakata 573-1010, Osaka, Japan
| | - Takashi Temma
- Department of Biofunctional Analysis, Graduate School of Pharmaceutical Sciences, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki 569-1094, Osaka, Japan; (M.F.); (A.M.); (N.K.)
| |
Collapse
|
3
|
Zhang X, Xiao J, Jiang M, Phillips CJC, Shi B. Thermogenesis and Energy Metabolism in Brown Adipose Tissue in Animals Experiencing Cold Stress. Int J Mol Sci 2025; 26:3233. [PMID: 40244078 PMCID: PMC11989373 DOI: 10.3390/ijms26073233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/29/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Cold exposure is a regulatory biological functions in animals. The interaction of thermogenesis and energy metabolism in brown adipose tissue (BAT) is important for metabolic regulation in cold stress. Brown adipocytes (BAs) produce uncoupling protein 1 (UCP1) in mitochondria, activating non-shivering thermogenesis (NST) by uncoupling fuel combustion from ATP production in response to cold stimuli. To elucidate the mechanisms underlying thermogenesis and energy metabolism in BAT under cold stress, we explored how cold exposure triggers the activation of BAT thermogenesis and regulates overall energy metabolism. First, we briefly outline the precursor composition and function of BA. Second, we explore the roles of the cAMP- protein kinase A (PKA) and adenosine monophosphate-activated protein kinase (AMPK) signaling pathways in thermogenesis and energy metabolism in BA during cold stress. Then, we analyze the mechanism by which BA regulates mitochondria homeostasis and energy balance during cold stress. This research reveals potential therapeutic targets, such as PKA, AMPK, UCP1 and PGC-1α, which can be used to develop innovative strategies for treating metabolic diseases. Furthermore, it provides theoretical support for optimizing cold stress response strategies, including the pharmacological activation of BAT and the genetic modulation of thermogenic pathways, to improve energy homeostasis in livestock.
Collapse
Affiliation(s)
- Xuekai Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.Z.); (M.J.); (B.S.)
| | - Jin Xiao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.Z.); (M.J.); (B.S.)
| | - Min Jiang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.Z.); (M.J.); (B.S.)
| | - Clive J. C. Phillips
- Curtin University Sustainability Policy (CUSP) Institute, Curtin University, Perth, WA 6845, Australia;
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51006 Tartu, Estonia
| | - Binlin Shi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.Z.); (M.J.); (B.S.)
| |
Collapse
|
4
|
Dongdem JT, Etornam AE, Beletaa S, Alidu I, Kotey H, Wezena CA. The β 3-Adrenergic Receptor: Structure, Physiopathology of Disease, and Emerging Therapeutic Potential. Adv Pharmacol Pharm Sci 2024; 2024:2005589. [PMID: 39640497 PMCID: PMC11620816 DOI: 10.1155/2024/2005589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
The discovery and characterization of the signal cascades of the β-adrenergic receptors have made it possible to effectively target the receptors for drug development. β-Adrenergic receptors are a class A rhodopsin type of G protein-coupled receptors (GPCRs) that are stimulated mainly by catecholamines and therefore mediate diverse effects of the parasympathetic nervous system in eliciting "fight or flight" type responses. They are detectable in several human tissues where they control a plethora of physiological processes and therefore contribute to the pathogenesis of several disease conditions. Given the relevance of the β-adrenergic receptor as a molecular target for many pathological conditions, this comprehensive review aims at providing an in-depth exploration of the recent advancements in β3-adrenergic receptor research. More importantly, we delve into the prospects of the β3-adrenergic receptor as a therapeutic target across a variety of clinical domains.
Collapse
Affiliation(s)
- Julius T. Dongdem
- Department of Chemical Pathology, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Axandrah E. Etornam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Solomon Beletaa
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Issah Alidu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Hassan Kotey
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Cletus A. Wezena
- Department of Microbiology, Faculty of Biosciences, University for Development Studies, Tamale, Northern Region, Ghana
| |
Collapse
|
5
|
Desai A, Loureiro ZY, DeSouza T, Yang Q, Solivan-Rivera J, Corvera S. cAMP driven UCP1 induction in human adipocytes requires ATGL-catalyzed lipolysis. Mol Metab 2024; 90:102051. [PMID: 39454826 PMCID: PMC11585812 DOI: 10.1016/j.molmet.2024.102051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
OBJECTIVE The uncoupling protein 1 (UCP1) is induced in brown or "beige" adipocytes through catecholamine-induced cAMP signaling, which activates diverse transcription factors. UCP1 expression can also be enhanced by PPARγ agonists such as rosiglitazone (Rsg). However, it is unclear whether this upregulation results from de-novo differentiation of beige adipocytes from progenitor cells, or from the induction of UCP1 in pre-existing adipocytes. To explore this, we employed human adipocytes differentiated from progenitor cells and examined their acute response to Rsg, to the adenylate-cyclase activator forskolin (Fsk), or to both simultaneously. METHODS Adipocytes generated from primary human progenitor cells were differentiated without exposure to PPARγ agonists, and treated for 3, 6 or 78 h to Fsk, to Rsg, or to both simultaneously. Bulk RNASeq, RNAScope, RT-PCR, CRISPR-Cas9 mediated knockout, oxygen consumption and western blotting were used to assess cellular responses. RESULTS UCP1 mRNA expression was induced within 3 h of exposure to either Rsg or Fsk, indicating that Rsg's effect is independent on additional adipocyte differentiation. Although Rsg and Fsk induced distinct overall transcriptional responses, both induced genes associated with calcium metabolism, lipid droplet assembly, and mitochondrial remodeling, denoting core features of human adipocyte beiging. Unexpectedly, we found that Fsk-induced UCP1 expression was reduced by approximately 80% following CRISPR-Cas9-mediated knockout of PNPLA2, the gene encoding the triglyceride lipase ATGL. As anticipated, ATGL knockout suppressed lipolysis; however, the associated suppression of UCP1 induction indicates that maximal cAMP-mediated UCP1 induction requires products of ATGL-catalyzed lipolysis. Supporting this, we observed that the reduction in Fsk-stimulated UCP1 induction caused by ATGL knockout was reversed by Rsg, implying that the role of lipolysis in this process is to generate natural PPARγ agonists. CONCLUSIONS UCP1 transcription is known to be stimulated by transcription factors activated downstream of cAMP-dependent protein kinases. Here we demonstrate that UCP1 transcription can also be acutely induced through PPARγ-activation. Moreover, both pathways are activated in human adipocytes in response to cAMP, synergistically inducing UCP1 expression. The stimulation of PPARγ in response to cAMP may result from the production of natural PPARγ activating ligands through ATGL-mediated lipolysis.
Collapse
Affiliation(s)
- Anand Desai
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Zinger Yang Loureiro
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA; Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Tiffany DeSouza
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Qin Yang
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA; Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Javier Solivan-Rivera
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA; Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA; Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
6
|
Sun C, Su J, Wang J, Ding K, Chen C. Lycium barbarum polysaccharide increases thermogenesis and energy metabolism through modulation of the gut microbiota to confer resistance to cold temperatures. FASEB J 2024; 38:e70010. [PMID: 39230621 DOI: 10.1096/fj.202400870r] [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: 04/17/2024] [Revised: 07/18/2024] [Accepted: 08/09/2024] [Indexed: 09/05/2024]
Abstract
Traditional Chinese medical literature contains numerous records of many traditional Chinese herbal medicines that exhibit efficacy in enhancing resistance to cold, yet there is a lack of scientific explanation. Lycium barbarum is among the herbal medicines that are explicitly documented to enhance resistance to cold in the "Ben Cao Gang Mu (Compendium of Materia Medica)". Herein, we investigated L. barbarum polysaccharide (LBP)-induced browning of inguinal white adipose tissue (iWAT), energy expenditure and thermogenic function in a long-term (4 months) treatment mouse model. LBP supplementation resulted in a significant reduction in weight and adipocyte size in iWAT, along with increased gut microbiota diversity. Specifically, the levels of Lachnospiraceae, Ruminococcaceae and Bacteroidaceae (short-chain fatty acid-producing bacteria) were elevated, leading to a higher level of short-chain fatty acids (SCFAs) in the caecal content. These effects subsequently triggered the release of glucagon-like peptide-1 (GLP-1) and activated the CREB/PGC1α signaling pathway in iWAT, thereby increasing energy expenditure and enhancing thermogenic function. The antibiotic treatment experiments confirmed that the LBP-mediated gut microbiota participated in the process of iWAT browning. In summary, our findings provide the first scientific explanation and mechanistic insights into the cold resistance of L. barbarum and identify potentially safe natural product supplements for individuals in alpine areas.
Collapse
Affiliation(s)
- Chuanxin Sun
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Juan Su
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Jiarui Wang
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Kan Ding
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, P.R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan, Guangdong, P.R. China
| | - Chang Chen
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, P.R. China
- Beijing Institute for Brain Disorders Capital Medical University, Beijing, P.R. China
| |
Collapse
|
7
|
Desai A, Yang Loureiro Z, DeSouza T, Yang Q, Solivan-Rivera J, Corvera S. PPARγ activation by lipolysis-generated ligands is required for cAMP dependent UCP1 induction in human thermogenic adipocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.10.607465. [PMID: 39211160 PMCID: PMC11360943 DOI: 10.1101/2024.08.10.607465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Objective The uncoupling protein 1 (UCP1) is induced in brown or "beige" adipocytes through catecholamine-induced cAMP signaling, which activates diverse transcription factors. UCP1 expression can also be enhanced by PPARγ agonists such as rosiglitazone (Rsg). However, it is unclear whether this upregulation results from de-novo differentiation of beige adipocytes from progenitor cells, or from the induction of UCP1 in pre-existing adipocytes. To explore this, we employed human adipocytes differentiated from progenitor cells and examined their acute response to Rsg, to the adenylate-cyclase activator forskolin (Fsk), or to both simultaneously. Methods Adipocytes generated from primary human progenitor cells were differentiated without exposure to PPARγ agonists, and treated for 3, 6 or 78 hours to Fsk, to Rsg, or to both simultaneously. Bulk RNASeq, RNAScope, RT-PCR, CRISPR-Cas9 mediated knockout, oxygen consumption and western blotting were used to assess cellular responses. Results UCP1 mRNA expression was induced within 3 hours of exposure to either Rsg or Fsk, indicating that Rsg's effect is independent on additional adipocyte differentiation. Although Rsg and Fsk induced distinct overall transcriptional responses, both induced genes associated with calcium metabolism, lipid droplet assembly, and mitochondrial remodeling, denoting core features of human adipocyte beiging. Unexpectedly, we found that Fsk-induced UCP1 expression was reduced by approximately 80% following CRISPR-Cas9-mediated knockout of PNPLA2 , the gene encoding the triglyceride lipase ATGL. As anticipated, ATGL knockout suppressed lipolysis; however, the associated suppression of UCP1 induction indicates that maximal cAMP-mediated UCP1 induction requires products of ATGL-catalyzed lipolysis. Supporting this, we observed that the reduction in Fsk-stimulated UCP1 induction caused by ATGL knockout was reversed by Rsg, implying that the role of lipolysis in this process is to generate natural PPARγ agonists. Conclusion UCP1 transcription is known to be stimulated by transcription factors activated downstream of cAMP-dependent protein kinases. Here we demonstrate that UCP1 transcription can also be acutely induced through PPARγ-activation. Moreover, both pathways are activated in human adipocytes in response to cAMP, synergistically inducing UCP1 expression. The stimulation of PPARγ in response to cAMP occurs as a result of the production of natural PPARγ activating ligands through ATGL-mediated lipolysis. GRAPHICAL ABSTRACT
Collapse
|
8
|
Lecoutre S, Maqdasy S, Rizo-Roca D, Renzi G, Vlassakev I, Alaeddine LM, Higos R, Jalkanen J, Zhong J, Zareifi DS, Frendo-Cumbo S, Massier L, Hodek O, Juvany M, Moritz T, de Castro Barbosa T, Omar-Hmeadi M, López-Yus M, Merabtene F, Abatan JB, Marcelin G, El Hachem EJ, Rouault C, Bergo MO, Petrus P, Zierath JR, Clément K, Krook A, Mejhert N, Rydén M. Reduced adipocyte glutaminase activity promotes energy expenditure and metabolic health. Nat Metab 2024; 6:1329-1346. [PMID: 39009762 PMCID: PMC11272588 DOI: 10.1038/s42255-024-01083-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/14/2024] [Indexed: 07/17/2024]
Abstract
Glutamine and glutamate are interconverted by several enzymes and alterations in this metabolic cycle are linked to cardiometabolic traits. Herein, we show that obesity-associated insulin resistance is characterized by decreased plasma and white adipose tissue glutamine-to-glutamate ratios. We couple these stoichiometric changes to perturbed fat cell glutaminase and glutamine synthase messenger RNA and protein abundance, which together promote glutaminolysis. In human white adipocytes, reductions in glutaminase activity promote aerobic glycolysis and mitochondrial oxidative capacity via increases in hypoxia-inducible factor 1α abundance, lactate levels and p38 mitogen-activated protein kinase signalling. Systemic glutaminase inhibition in male and female mice, or genetically in adipocytes of male mice, triggers the activation of thermogenic gene programs in inguinal adipocytes. Consequently, the knockout mice display higher energy expenditure and improved glucose tolerance compared to control littermates, even under high-fat diet conditions. Altogether, our findings highlight white adipocyte glutamine turnover as an important determinant of energy expenditure and metabolic health.
Collapse
Affiliation(s)
- Simon Lecoutre
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Salwan Maqdasy
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - David Rizo-Roca
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Gianluca Renzi
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Ivan Vlassakev
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Lynn M Alaeddine
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Romane Higos
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Jutta Jalkanen
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Jiawei Zhong
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Danae S Zareifi
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Scott Frendo-Cumbo
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Lucas Massier
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Ondrej Hodek
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Marta Juvany
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Thomas Moritz
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
- The Novo Nordisk Foundation Centre for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thais de Castro Barbosa
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Muhmmad Omar-Hmeadi
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Marta López-Yus
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, Zaragoza, Spain
- Instituto Aragonés de Ciencias de La Salud (IACS), Zaragoza, Spain
- Instituto de Investigación Sanitaria (IIS)-Aragón, Zaragoza, Spain
| | - Fatiha Merabtene
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Jimon Boniface Abatan
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Geneviève Marcelin
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Elie-Julien El Hachem
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Christine Rouault
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Martin O Bergo
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Paul Petrus
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Nutrition Department, Assistance Publique Hôpitaux de Paris, CRNH Ile-de-France, Pitié-Salpêtrière Hospital, Paris, France
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Niklas Mejhert
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden.
| | - Mikael Rydén
- Department of Medicine (Huddinge), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, Huddinge, Sweden.
| |
Collapse
|
9
|
Morales-Martínez M, Vega MI. p38 Molecular Targeting for Next-Generation Multiple Myeloma Therapy. Cancers (Basel) 2024; 16:256. [PMID: 38254747 PMCID: PMC10813990 DOI: 10.3390/cancers16020256] [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: 11/21/2023] [Revised: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Resistance to therapy and disease progression are the main causes of mortality in most cancers. In particular, the development of resistance is an important limitation affecting the efficacy of therapeutic alternatives for cancer, including chemotherapy, radiotherapy, and immunotherapy. Signaling pathways are largely responsible for the mechanisms of resistance to cancer treatment and progression, and multiple myeloma is no exception. p38 mitogen-activated protein kinase (p38) is downstream of several signaling pathways specific to treatment resistance and progression. Therefore, in recent years, developing therapeutic alternatives directed at p38 has been of great interest, in order to reverse chemotherapy resistance and prevent progression. In this review, we discuss recent findings on the role of p38, including recent advances in our understanding of its expression and activity as well as its isoforms, and its possible clinical role based on the mechanisms of resistance and progression in multiple myeloma.
Collapse
Affiliation(s)
- Mario Morales-Martínez
- Molecular Signal Pathway in Cancer Laboratory, UIMEO, Oncology Hospital, Siglo XXI National Medical Center, Mexican Institute of Social Security (IMSS), Mexico City 06720, Mexico
| | - Mario I. Vega
- Molecular Signal Pathway in Cancer Laboratory, UIMEO, Oncology Hospital, Siglo XXI National Medical Center, Mexican Institute of Social Security (IMSS), Mexico City 06720, Mexico
- Department of Medicine, Hematology-Oncology and Clinical Nutrition Division, Greater Los Angeles VA Healthcare Center, UCLA Medical Center, Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| |
Collapse
|
10
|
Matsumura Y, Osborne TF, Ito R, Takahashi H, Sakai J. β-Adrenergic Signal and Epigenomic Regulatory Process for Adaptive Thermogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1461:213-227. [PMID: 39289284 DOI: 10.1007/978-981-97-4584-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Activation of β-adrenergic (β-AR) signaling induces fight-or-flight stress responses which include enhancement of cardiopulmonary function, metabolic regulation, and muscle contraction. Classical dogma for β-AR signaling has dictated that the receptor-mediated response results in an acute and transient signal. However, more recent studies support more wide-ranging roles for β-AR signaling with long-term effects including cell differentiation that requires precisely timed and coordinated integration of many signaling pathways that culminate in precise epigenomic chromatin modifications. In this chapter, we discuss cold stress/β-AR signaling-induced epigenomic changes in adipose tissues that influence adaptive thermogenesis. We highlight recent studies showing dual roles for the histone demethylase JMJD1A as a mediator of both acute and chronic thermogenic responses to cold stress, in two distinct thermogenic tissues, and through two distinct molecular mechanisms. β-AR signaling not only functions through transient signals during acute stress responses but also relays a more sustained signal to long-term adaptation to environmental changes.
Collapse
Affiliation(s)
- Yoshihiro Matsumura
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Biochemistry and Metabolic Science, Akita University Graduate School of Medicine, Akita, Japan
| | - Timothy F Osborne
- Institute for Fundamental Biomedical Research Division of Endocrinology, Diabetes and Metabolism Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Ryo Ito
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroki Takahashi
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Juro Sakai
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan.
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
11
|
Wilson RA, Arivazhagan L, Ruiz HH, Zhou B, Qian K, Manigrasso MB, Bernadin R, Mangar K, Shekhtman A, Li H, Ramasamy R, Schmidt AM. Pharmacological antagonism of receptor for advanced glycation end products signaling promotes thermogenesis, healthful body mass and composition, and metabolism in mice. Obesity (Silver Spring) 2023; 31:1825-1843. [PMID: 37231626 PMCID: PMC10790363 DOI: 10.1002/oby.23774] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/14/2023] [Accepted: 02/28/2023] [Indexed: 05/27/2023]
Abstract
OBJECTIVE Optimal body mass and composition as well as metabolic fitness require tightly regulated and interconnected mechanisms across tissues. Disturbances in these regulatory networks tip the balance between metabolic health versus overweight and obesity and their complications. The authors previously demonstrated roles for the receptor for advanced glycation end products (RAGE) in obesity, as global- or adipocyte-specific deletion of Ager (the gene encoding RAGE) protected mice from high-fat diet-induced obesity and metabolic dysfunction. METHODS To explore translational strategies evoked by these observations, a small molecule antagonist of RAGE signaling, RAGE229, was administered to lean mice and mice with obesity undergoing diet-induced weight loss. Body mass and composition and whole body and adipose tissue metabolism were examined. RESULTS This study demonstrates that antagonism of RAGE signaling reduced body mass and adiposity and improved glucose, insulin, and lipid metabolism in lean male and female mice and in male mice with obesity undergoing weight loss. In adipose tissue and in human and mouse adipocytes, RAGE229 enhanced phosphorylation of protein kinase A substrates, which augmented lipolysis, mitochondrial function, and thermogenic programs. CONCLUSIONS Pharmacological antagonism of RAGE signaling is a potent strategy to optimize healthful body mass and composition and metabolic fitness.
Collapse
Affiliation(s)
- Robin A. Wilson
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Lakshmi Arivazhagan
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Henry H. Ruiz
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Boyan Zhou
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Kun Qian
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Michaele B. Manigrasso
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Rollanda Bernadin
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Kaamashri Mangar
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Alexander Shekhtman
- Department of Chemistry, State University of New York, Albany, New York, USA
| | - Huilin Li
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
12
|
Shaik Mohamed Sayed UF, Moshawih S, Goh HP, Kifli N, Gupta G, Singh SK, Chellappan DK, Dua K, Hermansyah A, Ser HL, Ming LC, Goh BH. Natural products as novel anti-obesity agents: insights into mechanisms of action and potential for therapeutic management. Front Pharmacol 2023; 14:1182937. [PMID: 37408757 PMCID: PMC10318930 DOI: 10.3389/fphar.2023.1182937] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/06/2023] [Indexed: 07/07/2023] Open
Abstract
Obesity affects more than 10% of the adult population globally. Despite the introduction of diverse medications aimed at combating fat accumulation and obesity, a significant number of these pharmaceutical interventions are linked to substantial occurrences of severe adverse events, occasionally leading to their withdrawal from the market. Natural products serve as attractive sources for anti-obesity agents as many of them can alter the host metabolic processes and maintain glucose homeostasis via metabolic and thermogenic stimulation, appetite regulation, pancreatic lipase and amylase inhibition, insulin sensitivity enhancing, adipogenesis inhibition and adipocyte apoptosis induction. In this review, we shed light on the biological processes that control energy balance and thermogenesis as well as metabolic pathways in white adipose tissue browning, we also highlight the anti-obesity potential of natural products with their mechanism of action. Based on previous findings, the crucial proteins and molecular pathways involved in adipose tissue browning and lipolysis induction are uncoupling protein-1, PR domain containing 16, and peroxisome proliferator-activated receptor-γ in addition to Sirtuin-1 and AMP-activated protein kinase pathway. Given that some phytochemicals can also lower proinflammatory substances like TNF-α, IL-6, and IL-1 secreted from adipose tissue and change the production of adipokines like leptin and adiponectin, which are important regulators of body weight, natural products represent a treasure trove for anti-obesity agents. In conclusion, conducting comprehensive research on natural products holds the potential to accelerate the development of an improved obesity management strategy characterized by heightened efficacy and reduced incidence of side effects.
Collapse
Affiliation(s)
| | - Said Moshawih
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Hui Poh Goh
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Nurolaini Kifli
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Andi Hermansyah
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas AirlanggaSurabaya, Indonesia
| | - Hooi Leng Ser
- School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Long Chiau Ming
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas AirlanggaSurabaya, Indonesia
- School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
Arianti R, Ágnes Vinnai B, Győry F, Guba A, Csősz É, Kristóf E, Fésüs L. Availability of abundant thiamine determines efficiency of thermogenic activation in human neck area derived adipocytes. J Nutr Biochem 2023:109385. [PMID: 37230255 DOI: 10.1016/j.jnutbio.2023.109385] [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: 06/29/2022] [Revised: 11/23/2022] [Accepted: 05/20/2023] [Indexed: 05/27/2023]
Abstract
Brown/beige adipocytes express uncoupling protein-1 (UCP1) that enables them to dissipate energy as heat. Systematic activation of this process can alleviate obesity. Human brown adipose tissues are interspersed in distinct anatomical regions including deep neck. We found that UCP1 enriched adipocytes differentiated from precursors of this depot highly expressed ThTr2 transporter of thiamine and consumed thiamine during thermogenic activation of these adipocytes by cAMP which mimics adrenergic stimulation. Inhibition of ThTr2 led to lower thiamine consumption with decreased proton leak respiration reflecting reduced uncoupling. In the absence of thiamine, cAMP-induced uncoupling was diminished but restored by thiamine addition reaching the highest levels at thiamine concentrations larger than present in human blood plasma. Thiamine is converted to thiamine pyrophosphate (TPP) in cells; the addition of TPP to permeabilized adipocytes increased uncoupling fueled by TPP-dependent pyruvate dehydrogenase. ThTr2 inhibition also hampered cAMP-dependent induction of UCP1, PGC1a, and other browning marker genes, and thermogenic induction of these genes was potentiated by thiamine in a concentration dependent manner. Our study reveals the importance of amply supplied thiamine during thermogenic activation in human adipocytes which provides TPP for TPP-dependent enzymes not fully saturated with this cofactor and by potentiating the induction of thermogenic genes.
Collapse
Affiliation(s)
- Rini Arianti
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, H-4032, Debrecen, Hungary
| | - Boglárka Ágnes Vinnai
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, H-4032, Debrecen, Hungary
| | - Ferenc Győry
- Department of Surgery, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary
| | - Andrea Guba
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, H-4032, Debrecen, Hungary
| | - Éva Csősz
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary
| | - Endre Kristóf
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary.
| | - László Fésüs
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary.
| |
Collapse
|
14
|
Xue S, Lee D, Berry DC. Thermogenic adipose tissue in energy regulation and metabolic health. Front Endocrinol (Lausanne) 2023; 14:1150059. [PMID: 37020585 PMCID: PMC10067564 DOI: 10.3389/fendo.2023.1150059] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
The ability to generate thermogenic fat could be a targeted therapy to thwart obesity and improve metabolic health. Brown and beige adipocytes are two types of thermogenic fat cells that regulate energy balance. Both adipocytes share common morphological, biochemical, and thermogenic properties. Yet, recent evidence suggests unique features exist between brown and beige adipocytes, such as their cellular origin and thermogenic regulatory processes. Beige adipocytes also appear highly plastic, responding to environmental stimuli and interconverting between beige and white adipocyte states. Additionally, beige adipocytes appear to be metabolically heterogenic and have substrate specificity. Nevertheless, obese and aged individuals cannot develop beige adipocytes in response to thermogenic fat-inducers, creating a key clinical hurdle to their therapeutic promise. Thus, elucidating the underlying developmental, molecular, and functional mechanisms that govern thermogenic fat cells will improve our understanding of systemic energy regulation and strive for new targeted therapies to generate thermogenic fat. This review will examine the recent advances in thermogenic fat biogenesis, molecular regulation, and the potential mechanisms for their failure.
Collapse
Affiliation(s)
| | | | - Daniel C. Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
15
|
Guarnieri AR, Benson TW, Tranter M. Calcium cycling as a mediator of thermogenic metabolism in adipose tissue. Mol Pharmacol 2022; 102:MOLPHARM-MR-2021-000465. [PMID: 35504660 PMCID: PMC9341262 DOI: 10.1124/molpharm.121.000465] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 11/22/2022] Open
Abstract
Canonical non-shivering thermogenesis (NST) in brown and beige fat relies on uncoupling protein 1 (UCP1)-mediated heat generation, although alternative mechanisms of NST have been identified, including sarcoplasmic reticulum (SR)-calcium cycling. Intracellular calcium is a crucial cell signaling molecule for which compartmentalization is tightly regulated, and the sarco-endoplasmic calcium ATPase (SERCA) actively pumps calcium from the cytosol into the SR. In this review, we discuss the capacity of SERCA-mediated calcium cycling as a significant mediator of thermogenesis in both brown and beige adipocytes. Here, we suggest two primary mechanisms of SR calcium mediated thermogenesis. The first mechanism is through direct uncoupling of the ATPase and calcium pump activity of SERCA, resulting in the energy of ATP catalysis being expended as heat in the absence of calcium transport. Regulins, a class of SR membrane proteins, act to decrease the calcium affinity of SERCA and uncouple the calcium transport function from ATPase activity, but remain largely unexplored in adipose tissue thermogenesis. A second mechanism is through futile cycling of SR calcium whereby SERCA-mediated SR calcium influx is equally offset by SR calcium efflux, resulting in ATP consumption without a net change in calcium compartmentalization. A fuller understanding of the functional and mechanistic role of calcium cycling as a mediator of adipose tissue thermogenesis and how manipulation of these pathways can be harnessed for therapeutic gain remains unexplored. Significance Statement Enhancing thermogenic metabolism in brown or beige adipose tissue may be of broad therapeutic utility to reduce obesity and metabolic syndrome. Canonical BAT-mediated thermogenesis occurs via uncoupling protein 1 (UCP1). However, UCP1-independent pathways of thermogenesis, such as sarcoplasmic (SR) calcium cycling, have also been identified, but the regulatory mechanisms and functional significance of these pathways remain largely unexplored. Thus, this mini-review discusses the state of the field with regard to calcium cycling as a thermogenic mediator in adipose tissue.
Collapse
Affiliation(s)
| | - Tyler W Benson
- University of Cincinnati College of Medicine, United States
| | | |
Collapse
|
16
|
Abstract
The role of β-adrenergic receptors (βARs) in adipose tissue to promote lipolysis and the release of fatty acids and nonshivering thermogenesis in brown fat has been studied for so many decades that one would think there is nothing left to discover. With the rediscovery of brown fat in humans and renewed interest in UCP1 and uncoupled mitochondrial respiration, it seems that a review of adipose tissue as an organ, pivotal observations, and the investigators who made them would be instructive to understanding where the field stands now. The discovery of the β3-adrenergic receptor was important for accurately defining the pharmacology of the adipocyte, while the clinical targeting of this receptor for obesity and metabolic disease has had its highs and lows. Many questions still remain about how βARs regulate adipocyte metabolism and the signaling molecules through which they do it.
Collapse
Affiliation(s)
- Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA;
| |
Collapse
|
17
|
Valentine JM, Ahmadian M, Keinan O, Abu-Odeh M, Zhao P, Zhou X, Keller MP, Gao H, Yu RT, Liddle C, Downes M, Zhang J, Lusis AJ, Attie AD, Evans RM, Rydén M, Saltiel AR. β3-Adrenergic receptor downregulation leads to adipocyte catecholamine resistance in obesity. J Clin Invest 2022; 132:e153357. [PMID: 34847077 PMCID: PMC8759781 DOI: 10.1172/jci153357] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
The dysregulation of energy homeostasis in obesity involves multihormone resistance. Although leptin and insulin resistance have been well characterized, catecholamine resistance remains largely unexplored. Murine β3-adrenergic receptor expression in adipocytes is orders of magnitude higher compared with that of other isoforms. While resistant to classical desensitization pathways, its mRNA (Adrb3) and protein expression are dramatically downregulated after ligand exposure (homologous desensitization). β3-Adrenergic receptor downregulation also occurs after high-fat diet feeding, concurrent with catecholamine resistance and elevated inflammation. This downregulation is recapitulated in vitro by TNF-α treatment (heterologous desensitization). Both homologous and heterologous desensitization of Adrb3 were triggered by induction of the pseudokinase TRIB1 downstream of the EPAC/RAP2A/PI-PLC pathway. TRIB1 in turn degraded the primary transcriptional activator of Adrb3, CEBPα. EPAC/RAP inhibition enhanced catecholamine-stimulated lipolysis and energy expenditure in obese mice. Moreover, adipose tissue expression of genes in this pathway correlated with body weight extremes in a cohort of genetically diverse mice and with BMI in 2 independent cohorts of humans. These data implicate a signaling axis that may explain reduced hormone-stimulated lipolysis in obesity and resistance to therapeutic interventions with β3-adrenergic receptor agonists.
Collapse
Affiliation(s)
| | | | | | | | | | - Xin Zhou
- Department of Pharmacology, Bioengineering, Chemistry, and Biochemistry, UCSD, San Diego, California, USA
| | - Mark P. Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hui Gao
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Ruth T. Yu
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, California, USA
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research and Sydney School of Medicine, University of Sydney, Westmead, New South Wales, Australia
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, California, USA
| | - Jin Zhang
- Department of Pharmacology, Bioengineering, Chemistry, and Biochemistry, UCSD, San Diego, California, USA
| | - Aldons J. Lusis
- Department of Microbiology, Immunology, and Molecular Genetics, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Alan D. Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ronald M. Evans
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, California, USA
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Alan R. Saltiel
- Department of Medicine and
- Department of Pharmacology, Bioengineering, Chemistry, and Biochemistry, UCSD, San Diego, California, USA
| |
Collapse
|
18
|
Mishra S, Sadagopan N, Dunkerly-Eyring B, Rodriguez S, Sarver DC, Ceddia RP, Murphy SA, Knutsdottir H, Jani VP, Ashok D, Oeing CU, O'Rourke B, Gangoiti JA, Sears DD, Wong GW, Collins S, Kass DA. Inhibition of phosphodiesterase type 9 reduces obesity and cardiometabolic syndrome in mice. J Clin Invest 2021; 131:148798. [PMID: 34618683 DOI: 10.1172/jci148798] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 09/16/2021] [Indexed: 12/16/2022] Open
Abstract
Central obesity with cardiometabolic syndrome (CMS) is a major global contributor to human disease, and effective therapies are needed. Here, we show that cyclic GMP-selective phosphodiesterase 9A inhibition (PDE9-I) in both male and ovariectomized female mice suppresses preestablished severe diet-induced obesity/CMS with or without superimposed mild cardiac pressure load. PDE9-I reduces total body, inguinal, hepatic, and myocardial fat; stimulates mitochondrial activity in brown and white fat; and improves CMS, without significantly altering activity or food intake. PDE9 localized at mitochondria, and its inhibition in vitro stimulated lipolysis in a PPARα-dependent manner and increased mitochondrial respiration in both adipocytes and myocytes. PPARα upregulation was required to achieve the lipolytic, antiobesity, and metabolic effects of PDE9-I. All these PDE9-I-induced changes were not observed in obese/CMS nonovariectomized females, indicating a strong sexual dimorphism. We found that PPARα chromatin binding was reoriented away from fat metabolism-regulating genes when stimulated in the presence of coactivated estrogen receptor-α, and this may underlie the dimorphism. These findings have translational relevance given that PDE9-I is already being studied in humans for indications including heart failure, and efficacy against obesity/CMS would enhance its therapeutic utility.
Collapse
Affiliation(s)
| | | | | | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ryan P Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Hildur Knutsdottir
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Vivek P Jani
- Division of Cardiology, Department of Medicine, and.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | - Jon A Gangoiti
- UCSD Biochemical Genetics and Metabolomics Laboratory and
| | - Dorothy D Sears
- Department of Medicine, UCSD, La Jolla, California, USA.,College of Health Solutions, Arizona State University, Phoenix, Arizona, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, and.,Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
19
|
Li Y, Zhang K, Liu J, Liu S, Nie C, Yan Y, Guan Y, Fan M, Qian H, Ying H, Wang L. Geniposide suppresses thermogenesis via regulating PKA catalytic subunit in adipocytes. Toxicology 2021; 464:153014. [PMID: 34718029 DOI: 10.1016/j.tox.2021.153014] [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: 09/09/2021] [Revised: 10/15/2021] [Accepted: 10/23/2021] [Indexed: 10/20/2022]
Abstract
Geniposide has been widely found to ameliorate many metabolic diseases. The recruitment and activation of brown/beige adipocytes are effective and promising methods for counteracting obesity and related diseases. However, the effect of geniposide on thermogenesis of adipocytes and its underlying mechanism have not yet been investigated. Here, we demonstrate that geniposide (25 mg/kg) reduces body temperature and cold tolerance of mice via suppressing thermogenic genes in interscapular brown adipose tissue (iBAT) and inguinal white adipose tissue (iWAT). Consistently, geniposide (20 mg/mL) suppresses thermogenic capacity of adipocytes (brown adipocytes and 3T3L1 preadipocyte cells) in vitro. Mechanistically, geniposide reduces the level of protein kinase A (PKA) catalytic subunit and further suppresses transcription activity and protein stability of uncoupling protein 1 (UCP1), leading to reduction of thermogenic capacity in adipocytes. Moreover, pharmacological PKA activation reverses geniposide-induced UCP1 inhibition, which indicated that geniposide suppresses thermogenesis of adipocytes via regulating PKA signaling. Together, our findings suggest that geniposide is an inhibitor of fat thermogenesis, establishing a novel function characteristic of geniposide.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - Kuiliang Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jinxin Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shengnan Liu
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China
| | - Chenzhipeng Nie
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Ying Yan
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China
| | - Yanming Guan
- China National Research Institute of Food and Fermentation Industries Co. Ltd, Beijing 100015, China
| | - Mingcong Fan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haifeng Qian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hao Ying
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China
| | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
20
|
Aburahma A, Pachhain S, Choudhury SR, Rana S, Phuntumart V, Larsen R, Sprague JE. Potential Contribution of the Intestinal Microbiome to Phenethylamine-Induced Hyperthermia. BRAIN, BEHAVIOR AND EVOLUTION 2021; 95:256-271. [PMID: 33472193 DOI: 10.1159/000512098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/05/2020] [Indexed: 12/26/2022]
Abstract
Phenethylamines (e.g., methamphetamine) are a common source of drug toxicity. Phenethylamine-induced hyperthermia (PIH) can activate a cascade of events that may result in rhabdomyolysis, coagulopathy, and even death. Here, we review recent evidence that suggests a potential link between the gut-brain axis and PIH. Within the preoptic area of the hypothalamus, phenethylamines lead to changes in catecholamine levels, that activate the sympathetic nervous system (SNS) and increase the peripheral levels of norepinephrine (NE), resulting in: (1) the loss of heat dissipation through α1 adrenergic receptor (α1-AR)-mediated vasoconstriction, (2) heat generation through β-AR activation and subsequent free fatty acid (FFA) activation of uncoupling proteins (UCPs) in brown and white adipose tissue, and (3) alteration of the gut microbiome and its link to the gut-brain axis. Recent studies have shown that phenethylamine derivatives can influence the composition of the gut microbiome and thus its metabolic potential. Phenethylamines increase the relative level of Proteuswhich has been linked to enhanced NE turnover. Bidirectional fecal microbial transplants (FMT) between PIH-tolerant and PIH-naïve rats demonstrated that the transplantation of gut microbiome can confer phenotypic hyperthermic and tolerant responses to phenethylamines. These phenethylamine-mediated changes in the gut microbiome were also associated with epigenetic changes in the mediators of thermogenesis. Given the significant role that the microbiome has been shown to play in the maintenance of body temperature, we outline current studies demonstrating the effects of phenethylamines on the gut microbiome and how these microbiome changes may mechanistically contribute to alterations in body temperature.
Collapse
Affiliation(s)
- Amal Aburahma
- The Ohio Attorney General's Center for the Future of Forensic Science, Bowling Green State University, Bowling Green, Ohio, USA
| | - Sudhan Pachhain
- The Department of Biological Sciences, Bowling Green State University, Bowling Green, Ohio, USA
| | - Sayantan Roy Choudhury
- The Department of Biological Sciences, Bowling Green State University, Bowling Green, Ohio, USA
| | - Srishti Rana
- The Department of Biological Sciences, Bowling Green State University, Bowling Green, Ohio, USA
| | - Vipa Phuntumart
- The Department of Biological Sciences, Bowling Green State University, Bowling Green, Ohio, USA
| | - Ray Larsen
- The Department of Biological Sciences, Bowling Green State University, Bowling Green, Ohio, USA
| | - Jon E Sprague
- The Ohio Attorney General's Center for the Future of Forensic Science, Bowling Green State University, Bowling Green, Ohio, USA,
| |
Collapse
|
21
|
Li H, Tang S. Baicalin attenuates diet-induced obesity partially through promoting thermogenesis in adipose tissue. Obes Res Clin Pract 2021; 15:485-490. [PMID: 34465552 DOI: 10.1016/j.orcp.2021.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 07/17/2021] [Accepted: 08/15/2021] [Indexed: 01/29/2023]
Abstract
BACKGROUND Adipose tissues have essential roles on energy homeostasis and the development of metabolic syndrome and obesity, they have become critical targets for treating obesity and metabolic disorders. Baicalin is a flavonoid that derived from the root of Scutellaria baicalensis, and it has been reported to take part in the regulation of adipocyte function. All these highlighted the potential of baicalin in the regulation of fat accumulation and obesity. Yet the impact of baicalin on thermogenic function of adipocytes remains to be deciphered. OBJECTIVE This study aims to explore the anti-obesity effects of baicalin. MATERIALS & METHODS The level of mRNA was detected by qRT-PCR and the protein expression level was examined by western blot. H&E staining was used for the observation of the structure of adipose tissue. Serum triglyceride and insulin levels were detected by commercial test kits. RESULTS Our data demonstrated that baicalin up-regulates the expression of UCP1 and PGC1a in a dose-dependent manner in vitro. Baicalin also increases ERK phosphorylation, and the increased expression of UCP1 and PGC1a in adipocytes could be inhibited by an ERK inhibitor, U0126. Moreover, dietary baicalin ameliorates high fat diet (HFD)-induced obesity without affecting food intake. In addition, dietary baicalin inhibits adipocyte hypertrophy and enhances thermogenic gene program in sWAT and intrascapular brown adipose tissue (iBAT) in vivo. DISCUSSION & CONCLUSION Baicalin prevents HFD-induced obesity partially through promoting adipocyte thermogenesis. Baicalin may be a promising compound against human obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Hongdi Li
- School of Sports Training, Wuhan Sports University, Wuhan 430079, China
| | - Shouyan Tang
- School of Sports Science, Lingnan Normal University, Zhanjiang 524033, China.
| |
Collapse
|
22
|
Zhang Z, Yang D, Xiang J, Zhou J, Cao H, Che Q, Bai Y, Guo J, Su Z. Non-shivering Thermogenesis Signalling Regulation and Potential Therapeutic Applications of Brown Adipose Tissue. Int J Biol Sci 2021; 17:2853-2870. [PMID: 34345212 PMCID: PMC8326120 DOI: 10.7150/ijbs.60354] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/23/2021] [Indexed: 12/25/2022] Open
Abstract
In mammals, thermogenic organs exist in the body that increase heat production and enhance energy regulation. Because brown adipose tissue (BAT) consumes energy and generates heat, increasing energy expenditure via BAT might be a potential strategy for new treatments for obesity and obesity-related diseases. Thermogenic differentiation affects normal adipose tissue generation, emphasizing the critical role that common transcriptional regulation factors might play in common characteristics and sources. An understanding of thermogenic differentiation and related factors could help in developing ways to improve obesity indirectly or directly through targeting of specific signalling pathways. Many studies have shown that the active components of various natural products promote thermogenesis through various signalling pathways. This article reviews recent major advances in this field, including those in the cyclic adenosine monophosphate-protein kinase A (cAMP-PKA), cyclic guanosine monophosphate-GMP-dependent protein kinase G (cGMP-AKT), AMP-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR), transforming growth factor-β/bone morphogenic protein (TGF-β/BMP), transient receptor potential (TRP), Wnt, nuclear factor-κ-light-chain-enhancer of activated B cells (NF-κΒ), Notch and Hedgehog (Hh) signalling pathways in brown and brown-like adipose tissue. To provide effective information for future research on weight-loss nutraceuticals or drugs, this review also highlights the natural products and their active ingredients that have been reported in recent years to affect thermogenesis and thus contribute to weight loss via the above signalling pathways.
Collapse
Affiliation(s)
- Zhengyan Zhang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Di Yang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Junwei Xiang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jingwen Zhou
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hua Cao
- Guangdong Cosmetics Engineering & Technology Research Center, School of Chemistry and Chemical Engneering, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd., Guangzhou 510663, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
23
|
Li J, Li E, Czepielewski RS, Chi J, Guo X, Han YH, Wang D, Wang L, Hu B, Dawes B, Jacobs C, Tenen D, Lin SJ, Lee B, Morris D, Tobias A, Randolph GJ, Cohen P, Tsai L, Rosen ED. Neurotensin is an anti-thermogenic peptide produced by lymphatic endothelial cells. Cell Metab 2021; 33:1449-1465.e6. [PMID: 34038712 PMCID: PMC8266750 DOI: 10.1016/j.cmet.2021.04.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/20/2021] [Accepted: 04/27/2021] [Indexed: 12/18/2022]
Abstract
The lymphatic vasculature plays important roles in the physiology of the organs in which it resides, though a clear mechanistic understanding of how this crosstalk is mediated is lacking. Here, we performed single-cell transcriptional profiling of human and mouse adipose tissue and found that lymphatic endothelial cells highly express neurotensin (NTS/Nts). Nts expression is reduced by cold and norepinephrine in an α-adrenergic-dependent manner, suggesting a role in adipose thermogenesis. Indeed, NTS treatment of brown adipose tissue explants reduced expression of thermogenic genes. Furthermore, adenoviral-mediated overexpression and knockdown or knockout of NTS in vivo reduced and enhanced cold tolerance, respectively, an effect that is mediated by NTSR2 and ERK signaling. Inhibition of NTSR2 promoted energy expenditure and improved metabolic function in obese mice. These data establish a link between adipose tissue lymphatics and adipocytes with potential therapeutic implications.
Collapse
Affiliation(s)
- Jin Li
- State Key Laboratory of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China; Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Erwei Li
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rafael S Czepielewski
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jingyi Chi
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Xiao Guo
- State Key Laboratory of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China
| | - Yong-Hyun Han
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daqing Wang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Luhong Wang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Bo Hu
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Brian Dawes
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Christopher Jacobs
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Danielle Tenen
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Samuel J Lin
- Division of Plastic Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Bernard Lee
- Division of Plastic Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Donald Morris
- Division of Plastic Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Adam Tobias
- Division of Plastic Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Linus Tsai
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute, Cambridge, MA, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
24
|
Pervin S, Reddy ST, Singh R. Novel Roles of Follistatin/Myostatin in Transforming Growth Factor-β Signaling and Adipose Browning: Potential for Therapeutic Intervention in Obesity Related Metabolic Disorders. Front Endocrinol (Lausanne) 2021; 12:653179. [PMID: 33897620 PMCID: PMC8062757 DOI: 10.3389/fendo.2021.653179] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is a global health problem and a major risk factor for several metabolic conditions including dyslipidemia, diabetes, insulin resistance and cardiovascular diseases. Obesity develops from chronic imbalance between energy intake and energy expenditure. Stimulation of cellular energy burning process has the potential to dissipate excess calories in the form of heat via the activation of uncoupling protein-1 (UCP1) in white and brown adipose tissues. Recent studies have shown that activation of transforming growth factor-β (TGF-β) signaling pathway significantly contributes to the development of obesity, and blockade or inhibition is reported to protect from obesity by promoting white adipose browning and increasing mitochondrial biogenesis. Identification of novel compounds that activate beige/brown adipose characteristics to burn surplus calories and reduce excess storage of fat are actively sought in the fight against obesity. In this review, we present recent developments in our understanding of key modulators of TGF-β signaling pathways including follistatin (FST) and myostatin (MST) in regulating adipose browning and brown adipose mass and activity. While MST is a key ligand for TGF-β family, FST can bind and regulate biological activity of several TGF-β superfamily members including activins, bone morphogenic proteins (BMP) and inhibins. Here, we review the literature supporting the critical roles for FST, MST and other proteins in modulating TGF-β signaling to influence beige and brown adipose characteristics. We further review the potential therapeutic utility of FST for the treatment of obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Shehla Pervin
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, CA, United States
| | - Srinivasa T. Reddy
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Rajan Singh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, CA, United States
- Department of Endocrinology, Men’s Health: Aging and Metabolism, Brigham and Women’s Hospital, Boston, MA, United States
- *Correspondence: Rajan Singh,
| |
Collapse
|
25
|
Watts AJ, Logan SM, Kübber-Heiss A, Posautz A, Stalder G, Painer J, Gasch K, Giroud S, Storey KB. Regulation of Peroxisome Proliferator-Activated Receptor Pathway During Torpor in the Garden Dormouse, Eliomys quercinus. Front Physiol 2020; 11:615025. [PMID: 33408645 PMCID: PMC7779809 DOI: 10.3389/fphys.2020.615025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/03/2020] [Indexed: 11/16/2022] Open
Abstract
Differential levels of n-6 and n-3 essential polyunsaturated fatty acids (PUFAs) are incorporated into the hibernator’s diet in the fall season preceding prolonged, multi-days bouts of torpor, known as hibernation. Peroxisome proliferator-activated receptor (PPAR) transcriptional activators bind lipids and regulate genes involved in fatty acid transport, beta-oxidation, ketogenesis, and insulin sensitivity; essential processes for survival during torpor. Thus, the DNA-binding activity of PPARα, PPARδ, PPARγ, as well as the levels of PPARγ coactivator 1α (PGC-1α) and L-fatty acid binding protein (L-FABP) were investigated in the hibernating garden dormouse (Eliomys quercinus). We found that dormice were hibernating in a similar way regardless of the n-6/n-3 PUFA diets fed to the animals during the fattening phase prior to hibernation. Further, metabolic rates and body mass loss during hibernation did not differ between dietary groups, despite marked differences in fatty acid profiles observed in white adipose tissue prior and at mid-hibernation. Overall, maintenance of PPAR DNA-binding activity was observed during torpor, and across three n-6/n-3 ratios, suggesting alternate mechanisms for the prioritization of lipid catabolism during torpor. Additionally, while no change was seen in L-FABP, significantly altered levels of PGC-1α were observed within the white adipose tissue and likely contributes to enhanced lipid metabolism when the diet favors n-6 PUFAs, i.e., high n-6/n-3 ratio, in both the torpid and euthermic state. Altogether, the maintenance of lipid metabolism during torpor makes it likely that consistent activity or levels of the investigated proteins are in aid of this metabolic profile.
Collapse
Affiliation(s)
| | | | - Anna Kübber-Heiss
- Department of Interdisciplinary Life Sciences, Research Institute of Wildlife Ecology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Annika Posautz
- Department of Interdisciplinary Life Sciences, Research Institute of Wildlife Ecology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Gabrielle Stalder
- Department of Interdisciplinary Life Sciences, Research Institute of Wildlife Ecology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Johanna Painer
- Department of Interdisciplinary Life Sciences, Research Institute of Wildlife Ecology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Kristina Gasch
- Department of Interdisciplinary Life Sciences, Research Institute of Wildlife Ecology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Sylvain Giroud
- Department of Interdisciplinary Life Sciences, Research Institute of Wildlife Ecology, University of Veterinary Medicine Vienna, Vienna, Austria
| | | |
Collapse
|
26
|
Hattori K, Wakatsuki H, Sakauchi C, Furutani S, Sugawara S, Hatta T, Natsume T, Ichijo H. β-adrenergic receptor signaling evokes the PKA-ASK axis in mature brown adipocytes. PLoS One 2020; 15:e0232645. [PMID: 33108364 PMCID: PMC7591029 DOI: 10.1371/journal.pone.0232645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/03/2020] [Indexed: 12/31/2022] Open
Abstract
Boosting energy expenditure by harnessing the activity of brown adipocytes is a promising strategy for combatting the global epidemic of obesity. Many studies have revealed that the β3-adrenergic receptor agonist is a potent activator of brown adipocytes, even in humans, and PKA and p38 MAPK have been demonstrated for regulating the transcription of a wide range of critical genes such as Ucp1. We previously revealed that the PKA-ASK1-p38 axis is activated in immature brown adipocytes and contributes to functional maturation. However, the downstream mechanisms of PKA that initiate the p38 MAPK cascade are still mostly unknown in mature brown adipocytes. Here, we identified the ASK family as a crucial signaling molecule bridging PKA and MAPK in mature brown adipocytes. Mechanistically, the phosphorylation of ASK1 at threonine 99 and serine 993 is critical in PKA-dependent ASK1 activation. Additionally, PKA also activates ASK2, which contributes to MAPK regulation. These lines of evidence provide new details for tailoring a βAR-dependent brown adipocyte activation strategy.
Collapse
Affiliation(s)
- Kazuki Hattori
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- * E-mail: (KH); (HI)
| | - Hiroaki Wakatsuki
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Chihiro Sakauchi
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shotaro Furutani
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Sho Sugawara
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomohisa Hatta
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Tohru Natsume
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Hidenori Ichijo
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- * E-mail: (KH); (HI)
| |
Collapse
|
27
|
Meyers K, López M, Ho J, Wills S, Rayalam S, Taval S. Lipocalin-2 deficiency may predispose to the progression of spontaneous age-related adiposity in mice. Sci Rep 2020; 10:14589. [PMID: 32883997 PMCID: PMC7471318 DOI: 10.1038/s41598-020-71249-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 08/13/2020] [Indexed: 01/10/2023] Open
Abstract
Lipocalin-2 (Lcn2) is an innate immune protein elevated by several orders of magnitude in various inflammatory conditions including aging and obesity. Recent studies have shown that Lcn2 is secreted by adipocytes in response to inflammation and is categorized as a new adipokine cross-linking innate immunity and metabolic disorders including obesity. However, the involvement of Lcn2 and its function during the progression of obesity is largely unknown. Recently, browning of white adipose tissue (WAT) has gained attention as a therapeutic strategy to combat obesity. Herein, we have shown that treatment of mature 3T3-L1 adipocytes with recombinant Lcn2 (rec-Lcn2) resulted in the up-regulation of thermogenic and beige/brown markers (UCP1, PRDM16, ZIC-1 and TBX1) and increased mitochondrial activity. Additionally, global Lcn2 genetic knockout (Lcn2KO) mice exhibited accelerated weight gain and visceral fat deposition with age, when compared to wild type (WT) mice. Taken together, both in vitro and in vivo studies suggest that Lcn2 is a naturally occurring adipokine, and may serve as an anti-obesity agent by upregulating the thermogenic markers resulting in the browning of WAT. Therefore, Lcn2 and its downstream signaling pathways could be a potential therapeutic target for obesity.
Collapse
Affiliation(s)
- Keya Meyers
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine - Georgia Campus, 625 Old Peachtree Road, Suwanee, GA, 30024, USA
| | - María López
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine - Georgia Campus, 625 Old Peachtree Road, Suwanee, GA, 30024, USA
| | - Joanna Ho
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine - Georgia Campus, 625 Old Peachtree Road, Suwanee, GA, 30024, USA
| | - Savannah Wills
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine - Georgia Campus, 625 Old Peachtree Road, Suwanee, GA, 30024, USA
| | - Srujana Rayalam
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine - Georgia Campus, 625 Old Peachtree Road, Suwanee, GA, 30024, USA. .,Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine, Room 3040, 625 Old Peachtree Road, Suwanee, GA, 30024, USA.
| | - Shashidharamurthy Taval
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine - Georgia Campus, 625 Old Peachtree Road, Suwanee, GA, 30024, USA. .,Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine, Room 3031, 625 Old Peachtree Road, Suwanee, GA, 30024, USA.
| |
Collapse
|
28
|
Vergnes L, Lin JY, Davies GR, Church CD, Reue K. Induction of UCP1 and thermogenesis by a small molecule via AKAP1/PKA modulation. J Biol Chem 2020; 295:15054-15069. [PMID: 32855239 DOI: 10.1074/jbc.ra120.013322] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/24/2020] [Indexed: 01/09/2023] Open
Abstract
Strategies to increase energy expenditure are an attractive approach to reduce excess fat storage and body weight to improve metabolic health. In mammals, uncoupling protein-1 (UCP1) in brown and beige adipocytes uncouples fatty acid oxidation from ATP generation in mitochondria and promotes energy dissipation as heat. We set out to identify small molecules that enhance UCP1 levels and activity using a high-throughput screen of nearly 12,000 compounds in mouse brown adipocytes. We identified a family of compounds that increase Ucp1 expression and mitochondrial activity (including un-coupled respiration) in mouse brown adipocytes and human brown and white adipocytes. The mechanism of action may be through compound binding to A kinase anchoring protein (AKAP) 1, modulating its localization to mitochondria and its interaction with protein kinase A (PKA), a known node in the β-adrenergic signaling pathway. In mice, the hit compound increased body temperature, UCP1 protein levels, and thermogenic gene expression. Some of the compound effects on mitochondrial function were UCP1- or AKAP1-independent, suggesting compound effects on multiple nodes of energy regulation. Overall, our results highlight a role for AKAP1 in thermogenesis, uncoupled respiration, and regulation energy balance.
Collapse
Affiliation(s)
- Laurent Vergnes
- Department of Human Genetics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California USA.
| | - Jason Y Lin
- Department of Human Genetics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California USA
| | - Graeme R Davies
- Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Christopher D Church
- Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California USA; Department of Medicine, and Molecular Biology Institute, University of California-Los Angeles, Los Angeles, California USA
| |
Collapse
|
29
|
Wei G, Sun H, Liu JL, Dong K, Liu J, Zhang M. Indirubin, a small molecular deriving from connectivity map (CMAP) screening, ameliorates obesity-induced metabolic dysfunction by enhancing brown adipose thermogenesis and white adipose browning. Nutr Metab (Lond) 2020; 17:21. [PMID: 32190098 PMCID: PMC7076951 DOI: 10.1186/s12986-020-00440-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 03/06/2020] [Indexed: 02/11/2023] Open
Abstract
Background Obesity occurs when the body’s energy intake is constantly greater than its energy consumption and the pharmacological enhancing the activity of brown adipose tissue (BAT) and (or) browning of white adipose tissue (WAT) has been considered promising strategies to treat obesity. Methods In this study, we took a multi-pronged approach to screen UCP1 activators, including in silico predictions, in vitro assays, as well as in vivo experiments. Results Base on Connectivity MAP (CMAP) screening, we obtained multiple drugs that possess a remarkably correlating gene expression pattern to that of enhancing activity in BAT and (or) sWAT signature. Particularly, we focused on a previously unreported drug-indirubin, a compound obtained from the Indigo plant, which is now mainly used for the treatment of chronic myelogenous leukemia (CML). In the current study, our results shown that indirubin could enhance the BAT activity, as evidenced by up-regulated Ucp1 expression and enhanced mitochondrial respiratory function in vitro cellular model. Furthermore, indirubin treatment restrained high-fat diet (HFD)-induced body weight gain, improved glucose homeostasis and ameliorated hepatic steatosis which were associated with the increase of energy expenditure in the mice model. Moreover, we revealed that indirubin treatment increased BAT activity by promoting thermogenesis and mitochondrial biogenesis in BAT and induced browning of subcutaneous inguinal white adipose tissue (sWAT) of mice under HFD. Besides, our results indicated that indirubin induced UCP1 expression in brown adipocytes, at least in part, via activation of PKA and p38MAPK signaling pathways. Conclusions Our results clearly show that as an effective BAT (as well as beige cells) activator, indirubin may have a protective effect on the prevention and treatment of obesity and its complications.
Collapse
Affiliation(s)
- Gang Wei
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032 People's Republic of China
| | - Honglin Sun
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032 People's Republic of China
| | - Jun-Li Liu
- 2Henan Key Laboratory of Neurorestoratology, Henan International Joint Laboratory of Neurorestoratology for Senile Dementia, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100 Henan Province People's Republic of China
| | - Kai Dong
- 3Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003 People's Republic of China
| | - Junli Liu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032 People's Republic of China
| | - Min Zhang
- 4Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, 200030 People's Republic of China
| |
Collapse
|
30
|
Hu J, Wang Z, Tan BK, Christian M. Dietary polyphenols turn fat “brown”: A narrative review of the possible mechanisms. Trends Food Sci Technol 2020. [DOI: 10.1016/j.tifs.2020.01.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
31
|
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: 33] [Impact Index Per Article: 6.6] [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
|
32
|
Identification of key candidate genes and molecular pathways in white fat browning: an anti-obesity drug discovery based on computational biology. Hum Genomics 2019; 13:55. [PMID: 31699147 PMCID: PMC6836481 DOI: 10.1186/s40246-019-0239-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/25/2019] [Indexed: 01/16/2023] Open
Abstract
Background Obesity—with its increased risk of obesity-associated metabolic diseases—has become one of the greatest public health epidemics of the twenty-first century in affluent countries. To date, there are no ideal drugs for treating obesity. Studies have shown that activation of brown adipose tissue (BAT) can promote energy consumption and inhibit obesity, which makes browning of white adipose tissue (WAT) a potential therapeutic target for obesity. Our objective was to identify genes and molecular pathways associated with WAT and the activation of BAT to WAT browning, by using publicly available data and computational tools; this knowledge might help in targeting relevant signaling pathways for treating obesity and other related metabolic diseases. Results In this study, we used text mining to find out genes related to brown fat and white fat browning. Combined with biological process and pathway analysis in GeneCodis and protein-protein interaction analysis by using STRING and Cytoscape, a list of high priority target genes was developed. The Human Protein Atlas was used to analyze protein expression. Candidate drugs were derived on the basis of the drug-gene interaction analysis of the final genes. Our study identified 18 genes representing 6 different pathways, targetable by a total of 33 drugs as possible drug treatments. The final list included 18 peroxisome proliferator-activated receptor gamma (PPAR-γ) agonists, 4 beta 3 adrenoceptor (β3-AR) agonists, 1 insulin sensitizer, 3 insulins, 6 lipase clearing factor stimulants and other drugs. Conclusions Drug discovery using in silico text mining, pathway, and protein-protein interaction analysis tools may be a method of exploring drugs targeting the activation of brown fat or white fat browning, which provides a basis for the development of novel targeted therapies as potential treatments for obesity and related metabolic diseases.
Collapse
|
33
|
Reverte-Salisa L, Sanyal A, Pfeifer A. Role of cAMP and cGMP Signaling in Brown Fat. Handb Exp Pharmacol 2019; 251:161-182. [PMID: 29633180 DOI: 10.1007/164_2018_117] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cold-induced activation of brown adipose tissue (BAT) is mediated by norepinephrine and adenosine that are released during sympathetic nerve activation. Both signaling molecules induce an increase in intracellular levels of 3',5'-cyclic adenosine monophosphate (cAMP) in murine and human BAT. In brown adipocytes, cAMP plays a central role, because it activates lipolysis, glucose uptake, and thermogenesis. Another well-studied intracellular second messenger is 3',5'-cyclic guanosine monophosphate (cGMP), which closely resembles cAMP. Several studies have shown that intact cGMP signaling is essential for normal adipogenic differentiation and BAT-mediated thermogenesis in mice. This chapter highlights recent observations, demonstrating the physiological significance of cyclic nucleotide signaling in BAT as well as their potential to induce browning of white adipose tissue (WAT) in mice and humans.
Collapse
Affiliation(s)
- Laia Reverte-Salisa
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Abhishek Sanyal
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
34
|
Role of epicardial adipose tissue NPR-C in acute coronary syndrome. Atherosclerosis 2019; 286:79-87. [DOI: 10.1016/j.atherosclerosis.2019.05.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/28/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022]
|
35
|
Feng Z, Wei Y, Zhang Y, Qiu Y, Liu X, Su L, Liang N, Yin H, Ding Q. Identification of a rhodanine derivative BML-260 as a potent stimulator of UCP1 expression. Theranostics 2019; 9:3501-3514. [PMID: 31281493 PMCID: PMC6587176 DOI: 10.7150/thno.31951] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 04/14/2019] [Indexed: 01/23/2023] Open
Abstract
Identification of proper agents to increase or activate UCP1+ cells in adipose tissues remains a potent therapeutic strategy to combat obesity. Screening systems for UCP1 activators have been previously established and allow for unbiased discovery of effective compound(s). Methods: A previously established Ucp1-2A-GFP reporter system was applied to a chemical library containing 33 phosphatase inhibitors. Compounds that can significantly activate UCP1 expression were further tested in vivo in mouse adipose tissues. Possible underlying mechanism was explored via RNA profiling, CMAP analysis, CRISPR targeting as well as inhibitor treatments. Results: We identified BML-260, a known potent inhibitor of the dual-specific phosphatase JSP-1, that significantly increased UCP1 expression in both brown and white adipocytes. BML-260 treatment also activated oxidative phosphorylation genes, increased mitochondrial activity as well as heat generation in vitro and in vivo. Mechanistic studies revealed that effect of BML-260 on adipocytes was partly through activated CREB, STAT3 and PPAR signaling pathways, and was unexpectedly JSP-1 independent. Conclusion: The rhodanine derivate BML-260 was previously identified to be a JSP-1 inhibitor, and thus was proposed to treat inflammatory and proliferative disorders associated with dysfunctional JNK signaling. This work provides evidences that BML-260 can also exert a JSP-1-independent effect in activating UCP1 and thermogenesis in adipocytes, and be potentially applied to treat obesity.
Collapse
|
36
|
Higareda-Almaraz JC, Karbiener M, Giroud M, Pauler FM, Gerhalter T, Herzig S, Scheideler M. Norepinephrine triggers an immediate-early regulatory network response in primary human white adipocytes. BMC Genomics 2018; 19:794. [PMID: 30390616 PMCID: PMC6215669 DOI: 10.1186/s12864-018-5173-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/16/2018] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Norepinephrine (NE) signaling has a key role in white adipose tissue (WAT) functions, including lipolysis, free fatty acid liberation and, under certain conditions, conversion of white into brite (brown-in-white) adipocytes. However, acute effects of NE stimulation have not been described at the transcriptional network level. RESULTS We used RNA-seq to uncover a broad transcriptional response. The inference of protein-protein and protein-DNA interaction networks allowed us to identify a set of immediate-early genes (IEGs) with high betweenness, validating our approach and suggesting a hierarchical control of transcriptional regulation. In addition, we identified a transcriptional regulatory network with IEGs as master regulators, including HSF1 and NFIL3 as novel NE-induced IEG candidates. Moreover, a functional enrichment analysis and gene clustering into functional modules suggest a crosstalk between metabolic, signaling, and immune responses. CONCLUSIONS Altogether, our network biology approach explores for the first time the immediate-early systems level response of human adipocytes to acute sympathetic activation, thereby providing a first network basis of early cell fate programs and crosstalks between metabolic and transcriptional networks required for proper WAT function.
Collapse
Affiliation(s)
- Juan Carlos Higareda-Almaraz
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Metabolic Control, Medical Faculty, Technical University, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- NMR laboratory, Institute of Myology, Hopital Universitaire Pitie Salpetriere, Paris, France
| | - Michael Karbiener
- Department of Phoniatrics, ENT University Hospital, Medical University of Graz, Graz, Austria
| | - Maude Giroud
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Metabolic Control, Medical Faculty, Technical University, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Florian M. Pauler
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Present Address: Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Teresa Gerhalter
- Present Address: Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Metabolic Control, Medical Faculty, Technical University, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marcel Scheideler
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Metabolic Control, Medical Faculty, Technical University, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- NMR laboratory, Institute of Myology, Hopital Universitaire Pitie Salpetriere, Paris, France
| |
Collapse
|
37
|
Qiu Y, Sun Y, Xu D, Yang Y, Liu X, Wei Y, Chen Y, Feng Z, Li S, Reyad-Ul Ferdous M, Zhao Y, Xu H, Lao Y, Ding Q. Screening of FDA-approved drugs identifies sutent as a modulator of UCP1 expression in brown adipose tissue. EBioMedicine 2018; 37:344-355. [PMID: 30348622 PMCID: PMC6286640 DOI: 10.1016/j.ebiom.2018.10.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The pharmacological activation of thermogenesis in brown adipose tissue has long been considered promising strategies to treat obesity. However, identification of safe and effective agents remains a challenge. In this study, we addressed this challenge by developing a cellular system with a fluorescence readout, and applied in a high-throughput manner to screen for FDA-approved drugs that may activate endogenous UCP1 expression in adipocytes. METHODS We have generated a Ucp1-2A-GFP reporter mouse, in which GFP intensity serves as a surrogate of the endogenous expression level of UCP1 protein; and immortalized brown adipocytes were derived from this mouse model and applied in drug screening. Candidate drugs were further tested in mouse models either fed with normal chow or high fat diet to induce obesity. FINDINGS By using the cellular screening platform, we identified a group of FDA-approved drugs that can upregulate UCP1 expression in brown adipocyte, including previously known UCP1 activators and new candidate drugs. Further studies focusing on a previously unreported drug-sutent, revealed that sutent treatment could increase the energy expenditure and inhibit lipid synthesis in mouse adipose and liver tissues, resulting in improved metabolism and resistance to obesity. INTERPRETATION This study offered an easy-to-use cellular screening system for UCP1 activators, and provided a candidate list of FDA-approved drugs that can potentially treat obesity. Further study of these candidates may shed new light on the drug discovery towards obesity. FUND: National Key Research and Development Program and the Strategic Priority Research Program of the Chinese Academy of Sciences, etc. (250 words).
Collapse
Affiliation(s)
- Yan Qiu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Yingmin Sun
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Danqing Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Yuanyuan Yang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Xiaojian Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Yuda Wei
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Yanhao Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Zhuanghui Feng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Shuang Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Md Reyad-Ul Ferdous
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Yongxu Zhao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, PR China
| | - Yuanzhi Lao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, PR China.
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, PR China.
| |
Collapse
|
38
|
Zhang S, Cao H, Li Y, Jing Y, Liu S, Ye C, Wang H, Yu S, Peng C, Hui L, Wang YC, Zhang H, Guo F, Zhai Q, Wang H, Huang R, Zhang L, Jiang J, Liu W, Ying H. Metabolic benefits of inhibition of p38α in white adipose tissue in obesity. PLoS Biol 2018; 16:e2004225. [PMID: 29750781 PMCID: PMC5965899 DOI: 10.1371/journal.pbio.2004225] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 05/23/2018] [Accepted: 04/09/2018] [Indexed: 01/22/2023] Open
Abstract
p38 has long been known as a central mediator of protein kinase A (PKA) signaling in brown adipocytes, which positively regulate the transcription of uncoupling protein 1 (UCP-1). However, the physiological role of p38 in adipose tissues, especially the white adipose tissue (WAT), is largely unknown. Here, we show that mice lacking p38α in adipose tissues display a lean phenotype, improved metabolism, and resistance to diet-induced obesity. Surprisingly, ablation of p38α causes minimal effects on brown adipose tissue (BAT) in adult mice, as evident from undetectable changes in UCP-1 expression, mitochondrial function, body temperature (BT), and energy expenditure. In contrast, genetic ablation of p38α in adipose tissues not only markedly facilitates the browning in WAT upon cold stress but also prevents diet-induced obesity. Consistently, pharmaceutical inhibition of p38α remarkably enhances the browning of WAT and has metabolic benefits. Furthermore, our data suggest that p38α deficiency promotes white-to-beige adipocyte reprogramming in a cell-autonomous manner. Mechanistically, inhibition of p38α stimulates the UCP-1 transcription through PKA and its downstream cAMP-response element binding protein (CREB), which form a positive feedback loop that functions to reinforce the white-to-beige phenotypic switch during cold exposure. Together, our study reveals that inhibition of p38α is able to promote WAT browning and confer metabolic benefits. Our study also indicates that p38α in WAT represents an exciting pharmacological target to combat obesity and metabolic diseases. The functional brown adipose tissue (BAT) identified in human adults consists of not only classic brown adipocytes but also brown-like adipocytes (beige adipocytes), both of which are important for energy homeostasis. Due to the same ability to convert fat into heat as brown adipocytes, beige adipocytes have been considered as a novel pharmacological target to combat obesity. Growing evidence suggests that promoting the development and formation of beige adipocytes in white adipose tissue (WAT), also called the browning of WAT, is able to prevent diet-induced obesity and improve metabolism in rodents. Thus, understanding the molecular basis for the regulation of browning in WAT may help us to develop new strategies to counteract obesity and metabolic diseases. In this study, adipocyte-specific p38α knockout (Fp38αKO) mice are generated that display a lean phenotype, improved metabolism, and resistance to diet-induced obesity. Interestingly, we found that adipocyte p38α deficiency facilitates the browning in WAT. Then, we show that pharmaceutical inhibition of p38α enhances the browning in WAT and has metabolic benefits. We propose that inhibiting p38α in WAT, possibly combined with cold exposure, could constitute an exciting pharmacological target to combat obesity and metabolic diseases.
Collapse
Affiliation(s)
- Shengjie Zhang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hongchao Cao
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Li
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yanyan Jing
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shengnan Liu
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Cheng Ye
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hui Wang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shuxian Yu
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chengyuan Peng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lijian Hui
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-cheng Wang
- Shanghai Xuhui Central Hospital, Shanghai Clinical Center, Chinese Academy of Sciences, Shanghai, China
| | - Haibing Zhang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hui Wang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| | - Ruimin Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ling Zhang
- Department of Head and Neck Surgery, Fudan University Cancer Center and Department of Oncology, Fudan University, Shanghai Medical College, Shanghai, China
| | - Jingjing Jiang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Liu
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hao Ying
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
- * E-mail:
| |
Collapse
|
39
|
Markussen LK, Winther S, Wicksteed B, Hansen JB. GSK3 is a negative regulator of the thermogenic program in brown adipocytes. Sci Rep 2018; 8:3469. [PMID: 29472592 PMCID: PMC5823915 DOI: 10.1038/s41598-018-21795-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 02/09/2018] [Indexed: 01/23/2023] Open
Abstract
Brown adipose tissue is a promising therapeutic target in metabolic disorders due to its ability to dissipate energy and improve systemic insulin sensitivity and glucose homeostasis. β-Adrenergic stimulation of brown adipocytes leads to an increase in oxygen consumption and induction of a thermogenic gene program that includes uncoupling protein 1 (Ucp1) and fibroblast growth factor 21 (Fgf21). In kinase inhibitor screens, we have identified glycogen synthase kinase 3 (GSK3) as a negative regulator of basal and β-adrenergically stimulated Fgf21 expression in cultured brown adipocytes. In addition, inhibition of GSK3 also caused increased Ucp1 expression and oxygen consumption. β-Adrenergic stimulation triggered an inhibitory phosphorylation of GSK3 in a protein kinase A (PKA)-dependent manner. Mechanistically, inhibition of GSK3 activated the mitogen activated protein kinase (MAPK) kinase 3/6-p38 MAPK-activating transcription factor 2 signaling module. In summary, our data describe GSK3 as a novel negative regulator of β-adrenergic signaling in brown adipocytes.
Collapse
Affiliation(s)
- Lasse K Markussen
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark
| | - Sally Winther
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark
| | - Barton Wicksteed
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jacob B Hansen
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark.
| |
Collapse
|
40
|
Cong H, Zhong W, Wang Y, Ikuyama S, Fan B, Gu J. Pycnogenol® Induces Browning of White Adipose Tissue through the PKA Signaling Pathway in Apolipoprotein E-Deficient Mice. J Diabetes Res 2018; 2018:9713259. [PMID: 29577045 PMCID: PMC5822826 DOI: 10.1155/2018/9713259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/02/2017] [Accepted: 10/26/2017] [Indexed: 01/13/2023] Open
Abstract
Beige adipocytes in white adipose tissue (WAT) have received considerable recognition because of their potential protective effect against obesity. Pycnogenol (PYC), extracted from French maritime pine bark, has anti-inflammatory and antioxidant properties and can improve lipid profiles. However, the effect of PYC on obesity has never been explored. In this study, we investigated the effects of PYC on obesity and WAT browning in apolipoprotein E- (ApoE-) deficient mice. The results showed that PYC treatment clearly reversed body weight and the mass of eWAT gain resulting from a high-cholesterol and high-fat diet (HCD), but no difference in food intake. The morphology results showed that the size of the adipocytes in the PYC-treated mice was obviously smaller than that in the HCD-fed mice. Next, we found that PYC upregulated the expression of genes related to lipolysis (ATGL and HSL), while it decreased the mRNA level of PLIN1. PYC significantly increased the expression of UCP1 and other genes related to beige adipogenesis. Additionally, PYC increased the expression of proteins related to the protein kinase A (PKA) signaling pathway. The findings suggested that PYC decreased obesity by promoting lipolysis and WAT browning. Thus, PYC may be a novel therapeutic target for obesity.
Collapse
Affiliation(s)
- Huiying Cong
- Department of Endocrinology and Metabolism, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang 110001, China
- The Endocrine Institute and the Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang 110001, China
| | - Wenxia Zhong
- Department of Endocrinology and Metabolism, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang 110001, China
- The Endocrine Institute and the Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang 110001, China
| | - Yiying Wang
- Department of Endocrinology and Metabolism, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang 110001, China
- The Endocrine Institute and the Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang 110001, China
| | - Shoichiro Ikuyama
- Department of Clinical Investigation, Department of Diabetes, Endocrine and Rheumatic Diseases, Oita San-ai Medical Center, 1213 Ichi, Oita 870-1151, Japan
| | - Bin Fan
- Department of Neurology, Shengjing Hospital, China Medical University, No. 39 Huaxiang Road, Shenyang 110022, China
| | - Jianqiu Gu
- Department of Endocrinology and Metabolism, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang 110001, China
- The Endocrine Institute and the Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang 110001, China
| |
Collapse
|
41
|
Abstract
Brite/brown adipose tissue (BAT) is a thermogenic tissue able to dissipate energy via non-shivering thermogenesis. It is naturally activated by cold and has been demonstrated to increase thermogenic capacity, elevate energy expenditure, and to ultimately contribute to fat mass reduction. Thus, it emerges as novel therapeutic concept for pharmacological intervention in obesity and other metabolic disorders. Therefore, the comprehensive understanding of the regulatory network in thermogenic adipocytes is in demand.The surprising findings that (1) all human protein-coding genes make up not more than 2% of our genome, (2) organismal complexity goes well along with the percentage of nonprotein-coding sequences, and that (3) three quarters of our genome are pervasively transcribed, provide evidence that noncoding RNAs (ncRNAs) are not junk, but a significant and even predominant part of our transcriptome representing a treasure chest worth retrieving regulatory determinants in biological processes and diseases.In this chapter, the impact of regulatory small and long ncRNAs (lncRNAs) in particular microRNAs and lncRNAs on BAT formation and metabolic function and their involvement in physiological and pathological conditions has been reviewed.
Collapse
|
42
|
Hayashi M, Futawaka K, Koyama R, Fan Y, Matsushita M, Hirao A, Fukuda Y, Nushida A, Nezu S, Tagami T, Moriyama K. Effects of growth hormone on uncoupling protein 1 in white adipose tissues in obese mice. Growth Horm IGF Res 2017; 37:31-39. [PMID: 29111497 DOI: 10.1016/j.ghir.2017.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/17/2017] [Accepted: 10/22/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The transition of white adipocytes to beige cells (a phenomenon referred to as browning or beigeing) during obesity has been previously reported. Our study aimed to examine the mechanisms through which obesity induced by a high fat diet (HFD) affects uncoupling protein 1 (UCP1) expression via signal transduction and activator of transcription 5 (STAT5s). DESIGN Seven-week-old male C57BL/6J mice were fed a normal or HFD for 11weeks. Body weight, white adipose tissue weight, and blood lipid and glucose levels were measured. To unveil the molecular mechanisms of UCP1 expression in adipose tissue, we performed further studying 3T3-L1 cells using qRT-PCR. We also measured UCP1 promoter activity in the TSA201 cell line using a dual luciferase assay. In addition, we analyzed the predicted consensus sequences for STAT5 binding in the UCP1 promoter region. RESULTS Mice fed an HFD had higher body weight and intra-abdominal adipose tissues weight and a higher expression of UCP1, GH receptor (GHR), STATs, suppressors of cytokine signaling (SOCSs), and cytokine-inducible SH2-containing protein (CISH) compared to control mice. In 3T3-L1 cell studies, GH induced phosphorylation of the STAT5, SOCSs, CISH and UCP1 expressions. UCP1 promoter activity was associated with constitutively active STAT5 in a dose-dependent manner. We confirmed functional STAT5 binding sites at -425, -279, and -178bp of the UCP1 promoter. CONCLUSION We suggest that endogenous GH induces UCP1 expression in adipose tissue via STAT5.
Collapse
Affiliation(s)
- Misa Hayashi
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Kumi Futawaka
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Rie Koyama
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Yue Fan
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Midori Matsushita
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Asuka Hirao
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Yuki Fukuda
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Ayako Nushida
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Syoko Nezu
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Tetsuya Tagami
- Clinical Research Institute for Endocrine and Metabolic Diseases, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
| | - Kenji Moriyama
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan; Clinical Research Institute for Endocrine and Metabolic Diseases, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan.
| |
Collapse
|
43
|
p38α MAPK antagonizing JNK to control the hepatic fat accumulation in pediatric patients onset intestinal failure. Cell Death Dis 2017; 8:e3110. [PMID: 29022907 PMCID: PMC5682685 DOI: 10.1038/cddis.2017.523] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 08/23/2017] [Accepted: 09/07/2017] [Indexed: 12/30/2022]
Abstract
The p38α mitogen-activated protein kinase (MAPK) has been related to gluconeogenesis and lipid metabolism. However, the roles and related mechanisms of p38α MAPK in intestinal failure (IF)-associated liver steatosis remained poor understood. Here, our experimental evidence suggested that p38α MAPK significantly suppressed the fat accumulation in livers of IF patients mainly through two mechanisms. On the one hand, p38α MAPK increased hepatic bile acid (BA) synthesis by upregulating the expression of the rate-limiting enzyme cholesterol 7-α-hydroxylase (CYP7A1) and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), which in turn activated the transcription of the CYP7A1. On the other hand, p38α MAPK promoted fatty acid (FA) β-oxidation via upregulating peroxisome proliferator-activated receptor alpha (PPARα) and its transcriptional target genes carnitine palmitoyltransferase 1A (CPT1A) and peroxisomal acyl-coenzyme aoxidase 1 (ACOX1). Dual luciferase assays indicated that p38α MAPK increased the transcription of PPARα, PGC-1α and CYP7A1 by upregulating their promoters’ activities. In addition, in vitro and in vivo assays indicated p38α MAPK negatively regulates the hepatic steatosis by controlling JNK activation. In conculsion, our findings demonstrate that hepatic p38α MAPK functions as a negative regulator of liver steatosis in maintaining BA synthesis and FAO by antagonizing the c-Jun N-terminal kinase (JNK).
Collapse
|
44
|
Shi F, Collins S. Second messenger signaling mechanisms of the brown adipocyte thermogenic program: an integrative perspective. Horm Mol Biol Clin Investig 2017; 31:/j/hmbci.ahead-of-print/hmbci-2017-0062/hmbci-2017-0062.xml. [PMID: 28949928 DOI: 10.1515/hmbci-2017-0062] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 08/29/2017] [Indexed: 01/19/2023]
Abstract
β-adrenergic receptors (βARs) are well established for conveying the signal from catecholamines to adipocytes. Acting through the second messenger cyclic adenosine monophosphate (cAMP) they stimulate lipolysis and also increase the activity of brown adipocytes and the 'browning' of adipocytes within white fat depots (so-called 'brite' or 'beige' adipocytes). Brown adipose tissue mitochondria are enriched with uncoupling protein 1 (UCP1), which is a regulated proton channel that allows the dissipation of chemical energy in the form of heat. The discovery of functional brown adipocytes in humans and inducible brown-like ('beige' or 'brite') adipocytes in rodents have suggested that recruitment and activation of these thermogenic adipocytes could be a promising strategy to increase energy expenditure for obesity therapy. More recently, the cardiac natriuretic peptides and their second messenger cyclic guanosine monophosphate (cGMP) have gained attention as a parallel signaling pathway in adipocytes, with some unique features. In this review, we begin with some important historical work that touches upon the regulation of brown adipocyte development and physiology. We then provide a synopsis of some recent advances in the signaling cascades from β-adrenergic agonists and natriuretic peptides to drive thermogenic gene expression in the adipocytes and how these two pathways converge at a number of unexpected points. Finally, moving from the physiologic hormonal signaling, we discuss yet another level of control downstream of these signals: the growing appreciation of the emerging roles of non-coding RNAs as important regulators of brown adipocyte formation and function. In this review, we discuss new developments in our understanding of the signaling mechanisms and factors including new secreted proteins and novel non-coding RNAs that control the function as well as the plasticity of the brown/beige adipose tissue as it responds to the energy needs and environmental conditions of the organism.
Collapse
|
45
|
Jiang C, Zhai M, Yan D, Li D, Li C, Zhang Y, Xiao L, Xiong D, Deng Q, Sun W. Dietary menthol-induced TRPM8 activation enhances WAT "browning" and ameliorates diet-induced obesity. Oncotarget 2017; 8:75114-75126. [PMID: 29088850 PMCID: PMC5650405 DOI: 10.18632/oncotarget.20540] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/29/2017] [Indexed: 12/21/2022] Open
Abstract
Beige adipocytes are a new type of recruitable brownish adipocytes, with highly mitochondrial membrane uncoupling protein 1 expression and thermogenesis. Beige adipocytes were found among white adipocytes, especially in subcutaneous white adipose tissue (sWAT). Therefore, beige adipocytes may be involved in the regulation of energy metabolism and fat deposition. Transient receptor potential melastatin 8 (TRPM8), a Ca2+-permeable non-selective cation channel, plays vital roles in the regulation of various cellular functions. It has been reported that TRPM8 activation enhanced the thermogenic function of brown adiposytes. However, the involvement of TRPM8 in the thermogenic function of WAT remains unexplored. Our data revealed that TRPM8 was expressed in mouse white adipocytes at mRNA, protein and functional levels. The mRNA expression of Trpm8 was significantly increased in the differentiated white adipocytes than pre-adipocytes. Moreover, activation of TRPM8 by menthol enhanced the expression of thermogenic genes in cultured white aidpocytes. And menthol-induced increases of the thermogenic genes in white adipocytes was inhibited by either KT5720 (a protein kinase A inhibitor) or BAPTA-AM. In addition, high fat diet (HFD)-induced obesity in mice was significantly recovered by co-treatment with menthol. Dietary menthol enhanced WAT “browning” and improved glucose metabolism in HFD-induced obesity mice as well. Therefore, we concluded that TRPM8 might be involved in WAT “browning” by increasing the expression levels of genes related to thermogenesis and energy metabolism. And dietary menthol could be a novel approach for combating human obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Changyu Jiang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The Affiliated Nanshan People's Hospital of Shenzhen University, Shenzhen Municipal Sixth People's Hospital, Shenzhen 518060, China
| | - Mingzhu Zhai
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria
| | - Dong Yan
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The Affiliated Nanshan People's Hospital of Shenzhen University, Shenzhen Municipal Sixth People's Hospital, Shenzhen 518060, China
| | - Da Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Chen Li
- Laboratory of Medicinal Plant, School of Basic Medicine, Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Hubei 442000, China
| | - Yonghong Zhang
- Laboratory of Medicinal Plant, School of Basic Medicine, Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Hubei 442000, China
| | - Lizu Xiao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The Affiliated Nanshan People's Hospital of Shenzhen University, Shenzhen Municipal Sixth People's Hospital, Shenzhen 518060, China
| | - Donglin Xiong
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The Affiliated Nanshan People's Hospital of Shenzhen University, Shenzhen Municipal Sixth People's Hospital, Shenzhen 518060, China
| | - Qiwen Deng
- Department of Infectious Diseases and Shenzhen Municipal Key Laboratory for Endogenous Infection, The Affiliated Nanshan People's Hospital of Shenzhen University, Shenzhen Municipal Sixth People's Hospital, Shenzhen 518060, China
| | - Wuping Sun
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The Affiliated Nanshan People's Hospital of Shenzhen University, Shenzhen Municipal Sixth People's Hospital, Shenzhen 518060, China
| |
Collapse
|
46
|
Ng R, Hussain NA, Zhang Q, Chang C, Li H, Fu Y, Cao L, Han W, Stunkel W, Xu F. miRNA-32 Drives Brown Fat Thermogenesis and Trans-activates Subcutaneous White Fat Browning in Mice. Cell Rep 2017; 19:1229-1246. [PMID: 28494871 PMCID: PMC5637386 DOI: 10.1016/j.celrep.2017.04.035] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 12/08/2016] [Accepted: 04/12/2017] [Indexed: 01/20/2023] Open
Abstract
Brown adipose tissue (BAT) activation and subcutaneous white fat browning are essential components of the thermogenic response to cold stimulus in mammals. microRNAs have been shown to regulate both processes in cis. Here, we identify miR-32 as a BAT-specific super-enhancer-associated miRNA in mice that is selectively expressed in BAT and further upregulated during cold exposure. Inhibiting miR-32 in vivo led to impaired cold tolerance, decreased BAT thermogenesis, and compromised white fat browning as a result of reduced serum FGF21 levels. Further examination showed that miR-32 directly represses its target gene Tob1, thereby activating p38 MAP kinase signaling to drive FGF21 expression and secretion from BAT. BAT-specific miR-32 overexpression led to increased BAT thermogenesis and serum FGF21 levels, which further promotes white fat browning in trans. Our results suggested miR-32 and Tob1 as modulators of FGF21 signaling that can be manipulated for therapeutic benefit against obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Raymond Ng
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A(∗)STAR), Singapore 117609, Singapore
| | - Nurul Attiqah Hussain
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A(∗)STAR), Singapore 117609, Singapore
| | - Qiongyi Zhang
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A(∗)STAR), Singapore 117609, Singapore
| | - Chengwei Chang
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A(∗)STAR), Singapore 117609, Singapore
| | - Hongyu Li
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, A(∗)STAR, Singapore 138667, Singapore
| | - Yanyun Fu
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, A(∗)STAR, Singapore 138667, Singapore
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Weiping Han
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, A(∗)STAR, Singapore 138667, Singapore; Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore
| | - Walter Stunkel
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A(∗)STAR), Singapore 117609, Singapore
| | - Feng Xu
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A(∗)STAR), Singapore 117609, Singapore; Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore.
| |
Collapse
|
47
|
Singh R, Braga M, Reddy ST, Lee SJ, Parveen M, Grijalva V, Vergnes L, Pervin S. Follistatin Targets Distinct Pathways To Promote Brown Adipocyte Characteristics in Brown and White Adipose Tissues. Endocrinology 2017; 158:1217-1230. [PMID: 28324027 PMCID: PMC5460830 DOI: 10.1210/en.2016-1607] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 01/11/2017] [Indexed: 12/14/2022]
Abstract
We previously demonstrated that Fst expression is highest in brown adipose tissue (BAT) and skeletal muscle, but is also present at substantial levels in epididymal and subcutaneous white adipose tissues (WATs). Fst promotes mouse brown preadipocyte differentiation and promotes browning during differentiation of mouse embryonic fibroblasts. Fst-transgenic (Fst-Tg) mice show substantial increases in circulating Fst levels and increased brown adipose mass. BAT of Fst-Tg mice had increased expression of brown adipose-associated markers including uncoupling protein 1 (UCP1), PRDM16, PGC-1α, and Glut4. WATs from Fst-Tg mice show upregulation of brown/beige adipose markers and significantly increased levels of phosphorylated p38 MAPK/ERK1/2 proteins compared with the wild-type (WT) mice. Pharmacological inhibition of pp38 MAPK/pERK1/2 pathway of recombinant mouse Fst (rFst) treated differentiating 3T3-L1 cells led to significant blockade of Fst-induced UCP1 protein expression. On the other hand, BAT from Fst-Tg mice or differentiating mouse BAT cells treated with rFst show dramatic increase in Myf5 protein levels as well as upregulation of Zic1 and Lhx8 gene expression. Myf5 levels were significantly downregulated in Fst knock-out embryos and small inhibitory RNA-mediated inhibition of Myf5 led to significant inhibition of UCP1, Lhx8, and Zic1 gene expression and significant blockade of Fst-induced induction of UCP1 protein expression in mouse BAT cells. Both interscapular BAT and WAT tissues from Fst-Tg mice display enhanced response to CL316,243 treatment and decreased expression of pSmad3 compared with the WT mice. Therefore, our results indicate that Fst promotes brown adipocyte characteristics in both WAT and BAT depots in vivo through distinct mechanisms.
Collapse
MESH Headings
- 3T3-L1 Cells
- Adipocytes, Brown/physiology
- Adipocytes, White/physiology
- Adipose Tissue, Brown/anatomy & histology
- Adipose Tissue, Brown/physiology
- Adipose Tissue, White/anatomy & histology
- Adipose Tissue, White/physiology
- Animals
- Cell Differentiation/genetics
- Cell Transdifferentiation/genetics
- Cells, Cultured
- Embryo, Mammalian
- Female
- Follistatin/blood
- Follistatin/genetics
- Follistatin/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Signal Transduction/genetics
- Thermogenesis/genetics
Collapse
Affiliation(s)
- Rajan Singh
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, California 90059; Departments of
- 2Obstetrics and Gynecology and
| | - Melissa Braga
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, California 90059; Departments of
| | - Srinivasa T. Reddy
- 2Obstetrics and Gynecology and
- Medicine, Molecular and Medical Pharmacology and
| | - Se-Jin Lee
- Johns Hopkins University School of Medicine, Department of Molecular Biology and Genetics, Baltimore, Maryland 21205
| | - Meher Parveen
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, California 90059; Departments of
| | | | - Laurent Vergnes
- Molecular Biology and Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Shehla Pervin
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, California 90059; Departments of
- 2Obstetrics and Gynecology and
| |
Collapse
|
48
|
Bargut TCL, Souza-Mello V, Aguila MB, Mandarim-de-Lacerda CA. Browning of white adipose tissue: lessons from experimental models. Horm Mol Biol Clin Investig 2017; 31:hmbci-2016-0051. [PMID: 28099124 DOI: 10.1515/hmbci-2016-0051] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 12/01/2016] [Indexed: 04/25/2024]
Abstract
Beige or brite (brown-in-white) adipocytes are present in white adipose tissue (WAT) and have a white fat-like phenotype that when stimulated acquires a brown fat-like phenotype, leading to increased thermogenesis. This phenomenon is known as browning and is more likely to occur in subcutaneous fat depots. Browning involves the expression of many transcription factors, such as PR domain containing 16 (PRDM16) and peroxisome proliferator-activated receptor (PPAR)-γ, and of uncoupling protein (UCP)-1, which is the hallmark of thermogenesis. Recent papers pointed that browning can occur in the WAT of humans, with beneficial metabolic effects. This fact indicates that these cells can be targeted to treat a range of diseases, with both pharmacological and nutritional activators. Pharmacological approaches to induce browning include the use of PPAR-α agonist, adrenergic receptor stimulation, thyroid hormone administration, irisin and FGF21 induction. Most of them act through the induction of PPAR-γ coactivator (PGC) 1-α and the consequent mitochondrial biogenesis and UCP1 induction. About the nutritional inducers, several compounds have been described with multiple mechanisms of action. Some of these activators include specific amino acids restriction, capsaicin, bile acids, Resveratrol, and retinoic acid. Besides that, some classes of lipids, as well as many plant extracts, have also been implicated in the browning of WAT. In conclusion, the discovery of browning in human WAT opens the possibility to target the adipose tissue to fight a range of diseases. Studies have arisen showing promising results and bringing new opportunities in thermogenesis and obesity control.
Collapse
Affiliation(s)
- Thereza Cristina Lonzetti Bargut
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratorio de Morfometria, Metabolismo e Doença Cardiovascular, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, 20551-030 Rio de Janeiro, Brazil, Phone (+55.21) 2868-8316, Fax: 2868-8033, E-mail:
| |
Collapse
|
49
|
Berry DC, Jiang Y, Arpke RW, Close EL, Uchida A, Reading D, Berglund ED, Kyba M, Graff JM. Cellular Aging Contributes to Failure of Cold-Induced Beige Adipocyte Formation in Old Mice and Humans. Cell Metab 2017; 25:166-181. [PMID: 27889388 PMCID: PMC5226893 DOI: 10.1016/j.cmet.2016.10.023] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/13/2016] [Accepted: 10/28/2016] [Indexed: 01/08/2023]
Abstract
Cold temperatures induce progenitor cells within white adipose tissue to form beige adipocytes that burn energy and generate heat; this is a potential anti-diabesity therapy. However, the potential to form cold-induced beige adipocytes declines with age. This creates a clinical roadblock to potential therapeutic use in older individuals, who constitute a large percentage of the obesity epidemic. Here we show that aging murine and human beige progenitor cells display a cellular aging, senescence-like phenotype that accounts for their age-dependent failure. Activating the senescence pathway, either genetically or pharmacologically, in young beige progenitors induces premature cellular senescence and blocks their potential to form cold-induced beige adipocytes. Conversely, genetically or pharmacologically reversing cellular aging by targeting the p38/MAPK-p16Ink4a pathway in aged mouse or human beige progenitor cells rejuvenates cold-induced beiging. This in turn increases glucose sensitivity. Collectively, these data indicate that anti-aging or senescence modalities could be a strategy to induce beiging, thereby improving metabolic health in aging humans.
Collapse
Affiliation(s)
- Daniel C Berry
- Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Yuwei Jiang
- Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Robert W Arpke
- Lillehei Heart Institute, University Minnesota, Minneapolis, MN 55455, USA; Department of Medicine, University Minnesota, Minneapolis, MN 55455, USA
| | - Elizabeth L Close
- Division of Metabolic Mechanisms of Disease in the Advanced Imaging Research Center and Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Aki Uchida
- Division of Metabolic Mechanisms of Disease in the Advanced Imaging Research Center and Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - David Reading
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eric D Berglund
- Division of Metabolic Mechanisms of Disease in the Advanced Imaging Research Center and Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Michael Kyba
- Lillehei Heart Institute, University Minnesota, Minneapolis, MN 55455, USA; Department of Pediatrics, University Minnesota, Minneapolis, MN 55455, USA
| | - Jonathan M Graff
- Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
50
|
Villarroya F, Peyrou M, Giralt M. Transcriptional regulation of the uncoupling protein-1 gene. Biochimie 2016; 134:86-92. [PMID: 27693079 DOI: 10.1016/j.biochi.2016.09.017] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/25/2016] [Indexed: 02/08/2023]
Abstract
Regulated transcription of the uncoupling protein-1 (UCP1) gene, and subsequent UCP1 protein synthesis, is a hallmark of the acquisition of the differentiated, thermogenically competent status of brown and beige/brite adipocytes, as well as of the responsiveness of brown and beige/brite adipocytes to adaptive regulation of thermogenic activity. The 5' non-coding region of the UCP1 gene contains regulatory elements that confer tissue specificity, differentiation dependence, and neuro-hormonal regulation to UCP1 gene transcription. Two main regions-a distal enhancer and a proximal promoter region-mediate transcriptional regulation through interactions with a plethora of transcription factors, including nuclear hormone receptors and cAMP-responsive transcription factors. Co-regulators, such as PGC-1α, play a pivotal role in the concerted regulation of UCP1 gene transcription. Multiple interactions of transcription factors and co-regulators at the promoter region of the UCP1 gene result in local chromatin remodeling, leading to activation and increased accessibility of RNA polymerase II and subsequent gene transcription. Moreover, a commonly occurring A-to-G polymorphism in close proximity to the UCP1 gene enhancer influences the extent of UCP1 gene transcription. Notably, it has been reported that specific aspects of obesity and associated metabolic diseases are associated with human population variability at this site. On another front, the unique properties of the UCP1 promoter region have been exploited to develop brown adipose tissue-specific gene delivery tools for experimental purposes.
Collapse
Affiliation(s)
- Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine, Institut de Biomedicina (IBUB), University of Barcelona, Barcelona, Catalonia, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain; Institut de Recerca Pediàtrica Sant Joan de Déu, Barcelona, Catalonia, Spain.
| | - Marion Peyrou
- Department of Biochemistry and Molecular Biomedicine, Institut de Biomedicina (IBUB), University of Barcelona, Barcelona, Catalonia, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain; Institut de Recerca Pediàtrica Sant Joan de Déu, Barcelona, Catalonia, Spain
| | - Marta Giralt
- Department of Biochemistry and Molecular Biomedicine, Institut de Biomedicina (IBUB), University of Barcelona, Barcelona, Catalonia, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain; Institut de Recerca Pediàtrica Sant Joan de Déu, Barcelona, Catalonia, Spain
| |
Collapse
|