1
|
Alpaslan Ağaçdiken A, Göktaş Z. Berberine-induced browning and energy metabolism: mechanisms and implications. PeerJ 2025; 13:e18924. [PMID: 39931072 PMCID: PMC11809318 DOI: 10.7717/peerj.18924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
Obesity has become a global pandemic. The approaches researched to prevent it include decreasing energy intake and/or enhancing energy expenditure. Therefore, research on brown adipose tissue is of great importance. Brown adipose tissue is characterized by its high mitochondrial content. Mitochondrial uncoupling protein 1 (UCP1) releases energy as heat instead of chemical energy. Thermogenesis increases energy expenditure. Berberine, a phytochemical widely used in Asian countries, has positive effects on body weight control. While the precise mechanisms behind this effect remain unclear, the adenosine monophosphate-activated protein kinase (AMPK) pathway is known to play a crucial role. Berberine activates AMPK through phosphorylation, significantly impacting brown adipose tissue by enhancing lipolytic activity and increasing the expression of UCP1, peroxisome proliferator-activated receptor γ-co-activator-1α (PGC1α), and PR domain containing 16 (PRDM16). While investigating the mechanism of action of berberine, both the AMPK pathway is being examined in more detail and alternative pathways are being explored. One such pathway is growth differentiation factor 15 (GDF15), known for its appetite-suppressing effect. Berberine's low stability and bioavailability, which are the main obstacles to its clinical use, have been improved through the development of nanotechnological methods. This review examines the potential mechanisms of berberine on browning and summarizes the methods developed to enhance its effect.
Collapse
Affiliation(s)
| | - Zeynep Göktaş
- Department of Nutrition and Dietetics, Hacettepe University, Ankara, Turkey
| |
Collapse
|
2
|
Longo M, Bishnu A, Risiglione P, Montava-Garriga L, Cuenco J, Sakamoto K, MacKintosh C, Ganley IG. Opposing roles for AMPK in regulating distinct mitophagy pathways. Mol Cell 2024; 84:4350-4367.e9. [PMID: 39532100 DOI: 10.1016/j.molcel.2024.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/31/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Mitophagy degrades damaged mitochondria, but we show here that it can also target functional mitochondria. This latter scenario occurs during programmed mitophagy and involves the mitophagy receptors NIX and BNIP3. Although AMP-activated protein kinase (AMPK), the energy-sensing protein kinase, can influence damaged-induced mitophagy, its role in programmed mitophagy is unclear. We found that AMPK directly inhibits NIX-dependent mitophagy by triggering 14-3-3-mediated sequestration of ULK1, via ULK1 phosphorylation at two sites: Ser556 and an additional identified site, Ser694. By contrast, AMPK activation increases Parkin phosphorylation and enhances the rate of depolarization-induced mitophagy, independently of ULK1. We show that this happens both in cultured cells and tissues in vivo, using the mito-QC mouse model. Our work unveils a mechanism whereby AMPK activation downregulates mitophagy of functional mitochondria but enhances that of dysfunctional/damaged ones.
Collapse
Affiliation(s)
- Marianna Longo
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| | - Aniketh Bishnu
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| | - Pierpaolo Risiglione
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| | - Lambert Montava-Garriga
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| | - Joyceline Cuenco
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Carol MacKintosh
- Division of Molecular Cell and Developmental Biology, University of Dundee, Dundee DD1 5EH, Scotland.
| | - Ian G Ganley
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland.
| |
Collapse
|
3
|
Ding L, Huwyler F, Long F, Yang W, Binz J, Wernlé K, Pfister M, Klug M, Balaz M, Ukropcova B, Ukropec J, Wu C, Wang T, Gao M, Clavien PA, Dutkowski P, Tibbitt MW, Wolfrum C. Glucose controls lipolysis through Golgi PtdIns4P-mediated regulation of ATGL. Nat Cell Biol 2024; 26:552-566. [PMID: 38561547 PMCID: PMC11021197 DOI: 10.1038/s41556-024-01386-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024]
Abstract
Metabolic crosstalk of the major nutrients glucose, amino acids and fatty acids (FAs) ensures systemic metabolic homeostasis. The coordination between the supply of glucose and FAs to meet various physiological demands is especially important as improper nutrient levels lead to metabolic disorders, such as diabetes and metabolic dysfunction-associated steatohepatitis (MASH). In response to the oscillations in blood glucose levels, lipolysis is thought to be mainly regulated hormonally to control FA liberation from lipid droplets by insulin, catecholamine and glucagon. However, whether general cell-intrinsic mechanisms exist to directly modulate lipolysis via glucose sensing remains largely unknown. Here we report the identification of such an intrinsic mechanism, which involves Golgi PtdIns4P-mediated regulation of adipose triglyceride lipase (ATGL)-driven lipolysis via intracellular glucose sensing. Mechanistically, depletion of intracellular glucose results in lower Golgi PtdIns4P levels, and thus reduced assembly of the E3 ligase complex CUL7FBXW8 in the Golgi apparatus. Decreased levels of the E3 ligase complex lead to reduced polyubiquitylation of ATGL in the Golgi and enhancement of ATGL-driven lipolysis. This cell-intrinsic mechanism regulates both the pool of intracellular FAs and their extracellular release to meet physiological demands during fasting and glucose deprivation. Moreover, genetic and pharmacological manipulation of the Golgi PtdIns4P-CUL7FBXW8-ATGL axis in mouse models of simple hepatic steatosis and MASH, as well as during ex vivo perfusion of a human steatotic liver graft leads to the amelioration of steatosis, suggesting that this pathway might be a promising target for metabolic dysfunction-associated steatotic liver disease and possibly MASH.
Collapse
Affiliation(s)
- Lianggong Ding
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Florian Huwyler
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, ETH Zürich, Zurich, Switzerland
| | - Fen Long
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Wu Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Jonas Binz
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, ETH Zürich, Zurich, Switzerland
| | - Kendra Wernlé
- Department of Surgery and Transplantation, University of Zurich, Zurich, Switzerland
- Wyss Zurich Translational Center, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Matthias Pfister
- Department of Surgery and Transplantation, University of Zurich, Zurich, Switzerland
- Wyss Zurich Translational Center, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Manuel Klug
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Miroslav Balaz
- Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Barbara Ukropcova
- Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jozef Ukropec
- Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Chunyan Wu
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Tongtong Wang
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Min Gao
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
- Department of Pharmacy, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pierre-Alain Clavien
- Department of Surgery and Transplantation, University of Zurich, Zurich, Switzerland
- Wyss Zurich Translational Center, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Philipp Dutkowski
- Department of Surgery and Transplantation, University of Zurich, Zurich, Switzerland
| | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, ETH Zürich, Zurich, Switzerland
- Wyss Zurich Translational Center, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland.
| |
Collapse
|
4
|
Efthymiou V, Ding L, Balaz M, Sun W, Balazova L, Straub LG, Dong H, Simon E, Ghosh A, Perdikari A, Keller S, Ghoshdastider U, Horvath C, Moser C, Hamilton B, Neubauer H, Wolfrum C. Inhibition of AXL receptor tyrosine kinase enhances brown adipose tissue functionality in mice. Nat Commun 2023; 14:4162. [PMID: 37443109 PMCID: PMC10344962 DOI: 10.1038/s41467-023-39715-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
The current obesity epidemic and high prevalence of metabolic diseases necessitate efficacious and safe treatments. Brown adipose tissue in this context is a promising target with the potential to increase energy expenditure, however no pharmacological treatments activating brown adipose tissue are currently available. Here, we identify AXL receptor tyrosine kinase as a regulator of adipose function. Pharmacological and genetic inhibition of AXL enhance thermogenic capacity of brown and white adipocytes, in vitro and in vivo. Mechanistically, these effects are mediated through inhibition of PI3K/AKT/PDE signaling pathway, resulting in induction of nuclear FOXO1 localization and increased intracellular cAMP levels via PDE3/4 inhibition and subsequent stimulation of the PKA-ATF2 pathway. In line with this, both constitutive Axl deletion as well as inducible adipocyte-specific Axl deletion protect animals from diet-induced obesity concomitant with increases in energy expenditure. Based on these data, we propose AXL receptor as a target for the treatment of obesity.
Collapse
Affiliation(s)
- Vissarion Efthymiou
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Joslin Diabetes Center, Section of Integrative Physiology and Metabolism, Research Division, Harvard Medical School, Boston, MA, USA
| | - Lianggong Ding
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Miroslav Balaz
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Laboratory of Cellular and Molecular Metabolism, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Wenfei Sun
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lucia Balazova
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Laboratory of Cellular and Molecular Metabolism, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Leon G Straub
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Institute of Child Nutrition, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Karlsruhe, Germany
| | - Hua Dong
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Eric Simon
- Department of Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Adhideb Ghosh
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Aliki Perdikari
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Svenja Keller
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Mechanisms of Inherited Kidney Diseases Group, Institute of Physiology, University of Zurich, 8057, Zurich, Switzerland
| | - Umesh Ghoshdastider
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Carla Horvath
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Caroline Moser
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Bradford Hamilton
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Heike Neubauer
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Christian Wolfrum
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland.
| |
Collapse
|
5
|
AlZaim I, Eid AH, Abd-Elrahman KS, El-Yazbi AF. Adipose Tissue Mitochondrial Dysfunction and Cardiometabolic Diseases: On the Search for Novel Molecular Targets. Biochem Pharmacol 2022; 206:115337. [DOI: 10.1016/j.bcp.2022.115337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
6
|
The New Role of AMP-Activated Protein Kinase in Regulating Fat Metabolism and Energy Expenditure in Adipose Tissue. Biomolecules 2021; 11:biom11121757. [PMID: 34944402 PMCID: PMC8698496 DOI: 10.3390/biom11121757] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is characterized by excessive accumulation of fat in the body, which is triggered by a body energy intake larger than body energy consumption. Due to complications such as cardiovascular diseases, type 2 diabetes (T2DM), obstructive pneumonia and arthritis, as well as high mortality, morbidity and economic cost, obesity has become a major health problem. The global prevalence of obesity, and its comorbidities is escalating at alarming rates, demanding the development of additional classes of therapeutics to reduce the burden of disease further. As a central energy sensor, the AMP-activated protein kinase (AMPK) has recently been elucidated to play a paramount role in fat synthesis and catabolism, especially in regulating the energy expenditure of brown/beige adipose tissue and the browning of white adipose tissue (WAT). This review discussed the role of AMPK in fat metabolism in adipose tissue, emphasizing its role in the energy expenditure of brown/beige adipose tissue and browning of WAT. A deeper understanding of the role of AMPK in regulating fat metabolism and energy expenditure can provide new insights into obesity research and treatment.
Collapse
|
7
|
Yi X, Wu P, Liu J, He S, Gong Y, Xiong J, Xu X, Li W. Candidate kinases for adipogenesis and osteoblastogenesis from human bone marrow mesenchymal stem cells. Mol Omics 2021; 17:790-795. [PMID: 34318850 DOI: 10.1039/d1mo00160d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Adipogenesis and osteoblastogenesis (adipo-osteoblastogenesis) are closely related processes involving with the phosphorylation of numerous cytoplasmic proteins and key transcription factors. Despite the recognition of the importance of protein phosphorylation in adipo-osteoblastocyte biology, relatively little is known about the specific kinases for adipo-osteoblastogenesis. Here, we constructed the comprehensive gene transcriptional landscapes of kinases at 3, 5, and 7 days during adipo-osteoblastogenesis from human bone marrow mesenchymal stem cells (hMSCs). We identified forty-four and eight significant DEGs (differentially expressed genes) separately for adipo-osteoblastogenesis. Five significant DEGs, namely CAMK2A, NEK10, PAK3, PRKG2, and PTK2B, were simultaneously shared by adipo-osteoblastogenic anecdotes. Using a lentivirus system, we confirmed that PTK2B (non-receptor protein tyrosine kinase 2 beta) simultaneously inhibited adipo-osteoblastogenesis through RNAi assays, and PRKG2 (protein kinase cGMP-dependent 2) facilitated adipogenesis and weakened osteoblastogenesis. The only certainty was that the identified candidate significant DEGs encoding kinases responsible for protein phosphorylation, especially PTK2B and PRKG2, were the potential molecular switches of cell fate determination for hMSCs. This study would provide novel study targets for hMSC differentiation and potential clues for the therapy of the adipo-osteoblastogenic balance-derived disorders.
Collapse
Affiliation(s)
- Xia Yi
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, 17 Lufeng Road, Jiujiang 332000, China.
| | - Ping Wu
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, 17 Lufeng Road, Jiujiang 332000, China.
| | - Jianyun Liu
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, 17 Lufeng Road, Jiujiang 332000, China.
| | - Shan He
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, 17 Lufeng Road, Jiujiang 332000, China.
| | - Ying Gong
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, 17 Lufeng Road, Jiujiang 332000, China.
| | - Jianjun Xiong
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, 17 Lufeng Road, Jiujiang 332000, China.
| | - Xiaoyuan Xu
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, 17 Lufeng Road, Jiujiang 332000, China.
| | - Weidong Li
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, 17 Lufeng Road, Jiujiang 332000, China.
| |
Collapse
|
8
|
van der Vaart JI, Boon MR, Houtkooper RH. The Role of AMPK Signaling in Brown Adipose Tissue Activation. Cells 2021; 10:cells10051122. [PMID: 34066631 PMCID: PMC8148517 DOI: 10.3390/cells10051122] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity is becoming a pandemic, and its prevalence is still increasing. Considering that obesity increases the risk of developing cardiometabolic diseases, research efforts are focusing on new ways to combat obesity. Brown adipose tissue (BAT) has emerged as a possible target to achieve this for its functional role in energy expenditure by means of increasing thermogenesis. An important metabolic sensor and regulator of whole-body energy balance is AMP-activated protein kinase (AMPK), and its role in energy metabolism is evident. This review highlights the mechanisms of BAT activation and investigates how AMPK can be used as a target for BAT activation. We review compounds and other factors that are able to activate AMPK and further discuss the therapeutic use of AMPK in BAT activation. Extensive research shows that AMPK can be activated by a number of different kinases, such as LKB1, CaMKK, but also small molecules, hormones, and metabolic stresses. AMPK is able to activate BAT by inducing adipogenesis, maintaining mitochondrial homeostasis and inducing browning in white adipose tissue. We conclude that, despite encouraging results, many uncertainties should be clarified before AMPK can be posed as a target for anti-obesity treatment via BAT activation.
Collapse
Affiliation(s)
- Jamie I. van der Vaart
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Mariëtte R. Boon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Leiden University Medical Center, Einthoven Laboratory for Experimental Vascular Medicine, 2333 ZA Leiden, The Netherlands
- Correspondence: (M.R.B.); (R.H.H.)
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
- Correspondence: (M.R.B.); (R.H.H.)
| |
Collapse
|
9
|
Rhein P, Desjardins EM, Rong P, Ahwazi D, Bonhoure N, Stolte J, Santos MD, Ovens AJ, Ehrlich AM, Sanchez Garcia JL, Ouyang Q, Yabut JM, Kjolby M, Membrez M, Jessen N, Oakhill JS, Treebak JT, Maire P, Scott JW, Sanders MJ, Descombes P, Chen S, Steinberg GR, Sakamoto K. Compound- and fiber type-selective requirement of AMPKγ3 for insulin-independent glucose uptake in skeletal muscle. Mol Metab 2021; 51:101228. [PMID: 33798773 PMCID: PMC8381060 DOI: 10.1016/j.molmet.2021.101228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 12/20/2022] Open
Abstract
Objective The metabolic master-switch AMP-activated protein kinase (AMPK) mediates insulin-independent glucose uptake in muscle and regulates the metabolic activity of brown and beige adipose tissue (BAT). The regulatory AMPKγ3 isoform is uniquely expressed in skeletal muscle and potentially in BAT. Herein, we investigated the role that AMPKγ3 plays in mediating skeletal muscle glucose uptake and whole-body glucose clearance in response to small-molecule activators that act on AMPK via distinct mechanisms. We also assessed whether γ3 plays a role in adipose thermogenesis and browning. Methods Global AMPKγ3 knockout (KO) mice were generated. A systematic whole-body, tissue, and molecular phenotyping linked to glucose homeostasis was performed in γ3 KO and wild-type (WT) mice. Glucose uptake in glycolytic and oxidative skeletal muscle ex vivo as well as blood glucose clearance in response to small molecule AMPK activators that target the nucleotide-binding domain of the γ subunit (AICAR) and allosteric drug and metabolite (ADaM) site located at the interface of the α and β subunit (991, MK-8722) were assessed. Oxygen consumption, thermography, and molecular phenotyping with a β3-adrenergic receptor agonist (CL-316,243) treatment were performed to assess BAT thermogenesis, characteristics, and function. Results Genetic ablation of γ3 did not affect body weight, body composition, physical activity, and parameters associated with glucose homeostasis under chow or high-fat diet. γ3 deficiency had no effect on fiber-type composition, mitochondrial content and components, or insulin-stimulated glucose uptake in skeletal muscle. Glycolytic muscles in γ3 KO mice showed a partial loss of AMPKα2 activity, which was associated with reduced levels of AMPKα2 and β2 subunit isoforms. Notably, γ3 deficiency resulted in a selective loss of AICAR-, but not MK-8722-induced blood glucose-lowering in vivo and glucose uptake specifically in glycolytic muscle ex vivo. We detected γ3 in BAT and found that it preferentially interacts with α2 and β2. We observed no differences in oxygen consumption, thermogenesis, morphology of BAT and inguinal white adipose tissue (iWAT), or markers of BAT activity between WT and γ3 KO mice. Conclusions These results demonstrate that γ3 plays a key role in mediating AICAR- but not ADaM site binding drug-stimulated blood glucose clearance and glucose uptake specifically in glycolytic skeletal muscle. We also showed that γ3 is dispensable for β3-adrenergic receptor agonist-induced thermogenesis and browning of iWAT. Loss of AMPKγ3 reduces glucose uptake in glycolytic skeletal muscle and whole-body glucose clearance with AMP-mimetic drug. γ3 is not required for muscle glucose uptake and whole-body glucose clearance with ADaM site-targeted allosteric activators. γ3 is present and forms a trimeric complex with α2 and β2 in brown adipose tissue. γ3 is dispensable for adipose thermogenesis and browning in response to a β3-adrenergic receptor agonist.
Collapse
Affiliation(s)
- Philipp Rhein
- Nestlé Research, Société des Produits Nestlé S.A., EPFL Innovation Park, Lausanne, 1015, Switzerland; School of Life Sciences, EPFL Innovation Park, Lausanne, 1015, Switzerland
| | - Eric M Desjardins
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, L8N3Z5, Canada; Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8N3Z5, Canada
| | - Ping Rong
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Danial Ahwazi
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Nicolas Bonhoure
- Nestlé Research, Société des Produits Nestlé S.A., EPFL Innovation Park, Lausanne, 1015, Switzerland
| | - Jens Stolte
- Nestlé Research, Société des Produits Nestlé S.A., EPFL Innovation Park, Lausanne, 1015, Switzerland
| | - Matthieu D Santos
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | - Ashley J Ovens
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Fitzroy, VIC, 3065, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, 3000, Australia
| | - Amy M Ehrlich
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - José L Sanchez Garcia
- Nestlé Research, Société des Produits Nestlé S.A., EPFL Innovation Park, Lausanne, 1015, Switzerland
| | - Qian Ouyang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Julian M Yabut
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, L8N3Z5, Canada; Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8N3Z5, Canada
| | - Mads Kjolby
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Clinical Pharmacology and Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Mathieu Membrez
- Nestlé Research, Société des Produits Nestlé S.A., EPFL Innovation Park, Lausanne, 1015, Switzerland
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Clinical Pharmacology and Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Fitzroy, VIC, 3065, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, 3000, Australia
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Pascal Maire
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | - John W Scott
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, 3000, Australia; Protein Chemistry and Metabolism Unit, St Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| | - Matthew J Sanders
- Nestlé Research, Société des Produits Nestlé S.A., EPFL Innovation Park, Lausanne, 1015, Switzerland
| | - Patrick Descombes
- Nestlé Research, Société des Produits Nestlé S.A., EPFL Innovation Park, Lausanne, 1015, Switzerland
| | - Shuai Chen
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Gregory R Steinberg
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, L8N3Z5, Canada; Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8N3Z5, Canada
| | - Kei Sakamoto
- Nestlé Research, Société des Produits Nestlé S.A., EPFL Innovation Park, Lausanne, 1015, Switzerland; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, 2200, Denmark.
| |
Collapse
|
10
|
Feeding brown fat: dietary phytochemicals targeting non-shivering thermogenesis to control body weight. Proc Nutr Soc 2020; 79:338-356. [PMID: 32290888 PMCID: PMC7663322 DOI: 10.1017/s0029665120006928] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Excessive adipose accumulation, which is the main driver for the development of secondary metabolic complications, has reached epidemic proportions and combined pharmaceutical, educational and nutritional approaches are required to reverse the current rise in global obesity prevalence rates. Brown adipose tissue (BAT) is a unique organ able to dissipate energy and thus a promising target to enhance BMR to counteract a positive energy balance. In addition, active BAT might support body weight maintenance after weight loss to prevent/reduce relapse. Natural products deliver valuable bioactive compounds that have historically helped to alleviate disease symptoms. Interest in recent years has focused on identifying nutritional constituents that are able to induce BAT activity and thereby enhance energy expenditure. This review provides a summary of selected dietary phytochemicals, including isoflavones, catechins, stilbenes, the flavonoids quercetin, luteolin and resveratrol as well as the alkaloids berberine and capsaicin. Most of the discussed phytochemicals act through distinct molecular pathways e.g. sympathetic nerve activation, AMP-kinase signalling, SIRT1 activity or stimulation of oestrogen receptors. Thus, it might be possible to utilise this multitude of pathways to co-activate BAT using a fine-tuned combination of foods or combined nutritional supplements.
Collapse
|
11
|
Buhlmann E, Horváth C, Houriet J, Kiehlmann E, Radtke J, Marcourt L, Wolfender JL, Wolfrum C, Schröder S. Puerariae lobatae root extracts and the regulation of brown fat activity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 64:153075. [PMID: 31476558 DOI: 10.1016/j.phymed.2019.153075] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/09/2019] [Accepted: 08/22/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Obesity is one of the major health problems worldwide. The induction of brown adipocyte formation and activity represents a promising therapeutic option by increasing energy expenditure. Asian herbs have the potential to treat obesity, however, pharmacological effects should be well documented at the molecular level first. HYPOTHESIS A novel hypothesis-driven screening approach identified the root of Pueraria montana var. lobata (Willd.) Sanjappa & Pradeep (PLR) to have potential effects on obesity by stimulating brown adipocytes. STUDY DESIGN This study explored the metabolic effects of PLR water extract (PLRE) in a high-fat diet-induced obesity mouse model and characterized its secondary metabolite composition. METHODS Animals were orally treated daily for two weeks and the bioactivity of PLRE evaluated by measuring various parameters including body weight, circulating metabolites, energy expenditure and insulin sensitivity. The chemical composition of the mains components was obtained by HPLC-MS-ELSD-PDA. Based on the dereplication results and semi-quantitative estimation, pure molecules were selected for tests on adipocytes in vitro. RESULTS PLRE induces brown adipocyte activity and triggers the formation of brown-like cells in inguinal fat tissue, weight loss, and improved glucose metabolism. These effects are primarily caused by cell-autonomous activation of brown adipocytes and not by autonomic nervous system regulation. Even though the analysis of PLRE revealed puerarin as the most abundant secondary metabolite, it showed no effect on brown adipocyte formation and function. Brown adipocyte activity was induced dose-dependently by two other isoflavones, daidzein, and genistein. Daidzein is present in a very small amount in PLRE, but various glycosidic isoflavones, including puerarin, may release daidzein after metabolism. CONCLUSION This approach demonstrated the positive effects of PLRE on a diet-induced obesity mouse model and provided clues on the mode of action of PLRE at the molecular level.
Collapse
Affiliation(s)
- Elisabeth Buhlmann
- HanseMerkur Center for Traditional Chinese Medicine at the University Medical Center Hamburg-Eppendorf, Breitenfelder Straße 15, D-20251 Hamburg, Germany
| | - Carla Horváth
- Swiss Federal Institute of Technology, ETH Zürich, Institute of Food Nutrition and Health, Schorenstr. 16, CH-8603 Schwerzenbach, Switzerland
| | - Joëlle Houriet
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Rue Michel Servet 1, 1206 Genève, Switzerland
| | - Elke Kiehlmann
- Swiss Federal Institute of Technology, ETH Zürich, Institute of Food Nutrition and Health, Schorenstr. 16, CH-8603 Schwerzenbach, Switzerland
| | - Janine Radtke
- HanseMerkur Center for Traditional Chinese Medicine at the University Medical Center Hamburg-Eppendorf, Breitenfelder Straße 15, D-20251 Hamburg, Germany
| | - Laurence Marcourt
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Rue Michel Servet 1, 1206 Genève, Switzerland
| | - Jean-Luc Wolfender
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Rue Michel Servet 1, 1206 Genève, Switzerland
| | - Christian Wolfrum
- Swiss Federal Institute of Technology, ETH Zürich, Institute of Food Nutrition and Health, Schorenstr. 16, CH-8603 Schwerzenbach, Switzerland
| | - Sven Schröder
- HanseMerkur Center for Traditional Chinese Medicine at the University Medical Center Hamburg-Eppendorf, Breitenfelder Straße 15, D-20251 Hamburg, Germany.
| |
Collapse
|
12
|
Abstract
The organs require oxygen and other types of nutrients (amino acids, sugars, and lipids) to function, the heart consuming large amounts of fatty acids for oxidation and adenosine triphosphate (ATP) generation.
Collapse
|
13
|
Brendle C, Stefan N, Stef I, Ripkens S, Soekler M, la Fougère C, Nikolaou K, Pfannenberg C. Impact of diverse chemotherapeutic agents and external factors on activation of brown adipose tissue in a large patient collective. Sci Rep 2019; 9:1901. [PMID: 30760750 PMCID: PMC6374459 DOI: 10.1038/s41598-018-37924-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/18/2018] [Indexed: 11/21/2022] Open
Abstract
Increased activity of brown adipose tissue (BAT) activity in adults is thought to prevent obesity. Therefore, regulators of BAT activity might serve as anti-obesity therapy in future, but are not investigated thoroughly up to now. In our study, we assessed retrospectively the association of BAT activity with several external factors and diverse chemotherapeutic and immunosuppressive agents in a collective of 702 patients. The patients underwent at least two clinically indicated PET/CT examinations in the course of different oncological and inflammatory diseases. BAT activity was identified according to predefined PET/CT criteria in all examinations. In multivariate analysis, the type of disease, the disease activity and the therapeutic regimen did not influence BAT activity. In contrast, sex and age were confirmed as independent factors for BAT activity. For the association of therapeutic agents with BAT activity, we examined 53 different disease-related agents, which were applied to patients without initial BAT activity between their PET/CT examinations. Out of these, cytarabine therapy was significantly associated with increased new onset of BAT activity. Cytarabine is a therapeutic agent for lymphoma patients. Further targeted studies might investigate the usefulness of Cytarabine serving as possible therapeutic approach against obesity via BAT regulation.
Collapse
Affiliation(s)
- Cornelia Brendle
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany. .,Diagnostic and Interventional Neuroradiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany.
| | - Norbert Stefan
- Endocrinology and Diabetology, Department of Internal Medicine, Eberhard Karls University, Otfried-Mueller-Straße 10, 72076, Tuebingen, Germany
| | - Irina Stef
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany
| | - Sabine Ripkens
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany
| | - Martin Soekler
- Oncology, Hematology, Clinical Immunology, Rheumatology and Pulmology, Department of Internal Medicine, Eberhard Karls University, Otfried-Mueller-Straße 10, 72076, Tuebingen, Germany
| | - Christian la Fougère
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University, Otfried-Mueller-Straße 14, 72076, Tuebingen, Germany
| | - Konstantin Nikolaou
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany
| | - Christina Pfannenberg
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany
| |
Collapse
|
14
|
Desjardins EM, Steinberg GR. Emerging Role of AMPK in Brown and Beige Adipose Tissue (BAT): Implications for Obesity, Insulin Resistance, and Type 2 Diabetes. Curr Diab Rep 2018; 18:80. [PMID: 30120579 DOI: 10.1007/s11892-018-1049-6] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The global prevalence of type 2 diabetes (T2D) is escalating at alarming rates, demanding the development of additional classes of therapeutics to further reduce the burden of disease. Recent studies have indicated that increasing the metabolic activity of brown and beige adipose tissue may represent a novel means to reduce circulating glucose and lipids in people with T2D. The AMP-activated protein kinase (AMPK) is a cellular energy sensor that has recently been demonstrated to be important in potentially regulating the metabolic activity of brown and beige adipose tissue. The goal of this review is to summarize recent work describing the role of AMPK in brown and beige adipose tissue, focusing on its role in adipogenesis and non-shivering thermogenesis. RECENT FINDINGS Ablation of AMPK in mouse adipocytes results in cold intolerance, a reduction in non-shivering thermogenesis in brown adipose tissue (BAT), and the development of non-alcoholic fatty liver disease (NAFLD) and insulin resistance; effects associated with a defect in mitochondrial specific autophagy (mitophagy) within BAT. The effects of a β3-adrenergic agonist on the induction of BAT thermogenesis and the browning of white adipose tissue (WAT) are also blunted in mice lacking adipose tissue AMPK. A specific AMPK activator, A-769662, also results in the activation of BAT and the browning of WAT, effects which may involve demethylation of the PR domain containing 16 (Prdm16) promoter region, which is important for BAT development. AMPK plays an important role in the development and maintenance of brown and beige adipose tissue. Adipose tissue AMPK is reduced in people with insulin resistance, consistent with findings that mice lacking adipocyte AMPK develop greater NAFLD and insulin resistance. These data suggest that pharmacologically targeting adipose tissue AMPK may represent a promising strategy to enhance energy expenditure and reduce circulating glucose and lipids, which may be effective for the treatment of NAFLD and T2D.
Collapse
Affiliation(s)
- Eric M Desjardins
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8N 3Z5, Canada
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8N 3Z5, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8N 3Z5, Canada.
| |
Collapse
|
15
|
A novel role for PTK2B in cultured beige adipocyte differentiation. Biochem Biophys Res Commun 2018; 501:851-857. [DOI: 10.1016/j.bbrc.2018.05.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 12/27/2022]
|
16
|
Grempler R, Wolff M, Simon E, Schmid R, Eisele C, Rieber K, Fischer E, Mettel S, Gabrielyan O, Delic D, Luippold G, Redeman N. Discovery and translation of a target engagement marker for AMP-activated protein kinase (AMPK). PLoS One 2018; 13:e0197849. [PMID: 29799853 PMCID: PMC5969744 DOI: 10.1371/journal.pone.0197849] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 05/09/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Activation of the AMP-activated protein kinase (AMPK) is an attractive approach for the treatment of type 2 diabetes. AMPK activation reduces glucose levels in animal models of type 2 diabetes by increasing glucose uptake in skeletal muscles and reducing hepatic glucose production. Furthermore, AMPK activation ameliorates hepatic steatosis in animal models. For the clinical development of AMPK activators it is essential to have a reliable target engagement marker for appropriate dose finding and to support proof of clinical principle. While the activation of AMPK by quantification of the phosphorylation of AMPK at Thr172 in target tissues can be assessed pre-clinically, this is not feasible in clinical studies. Therefore, we attempted to identify and translate a peripheral target engagement biomarker downstream of AMPK activation for clinical use in blood samples. METHODS For pharmacological activation of AMPK, two AMPK activators were synthesized (compound 1 and 2). A compound with structural similarities but no pharmacological effect on AMPK phosphorylation was synthesized as negative control (compound 3). Whole blood from healthy volunteers was incubated with an AMPK activator for up to 6 hours and mRNA sequencing was performed. Additionally, human PBMCs were isolated to evaluate Thr172-phosphorylation of AMPK in Western blots. In order to enable identification of translatable biomarker candidates, blood samples from HanWistar rats treated for two weeks with an AMPK activator were also subjected to mRNA sequencing. Furthermore, concentration-response curves for four biomarker candidates were recorded in human blood samples using Nanostring nCounter technology. Finally, ZDF rats were treated with increasing doses of compound 2 for five weeks to investigate the glucose-lowering efficacy. To investigate changes of mRNA expression of two selected biomarker candidates in this ZDF rat study, qRT-PCR was performed. RESULTS Pharmacological activation of AMPK in human PBMCs revealed an increase in Thr172-phosphorylation of AMPK, confirming target engagement in these blood cells. RNA sequencing of human blood samples identified 608 deregulated genes after AMPK activation. Additionally, AMPK activation led to deregulation of 367 genes in whole blood from HanWistar rats which mapped to the respective human genes. 22 genes out of the intersection of genes deregulated in both species are proposed as potential translatable target engagement biomarker candidates. The most prominent genes were transmembrane glycoprotein NMB (GPNMB, osteoactivin), calcium-binding protein A9 (S100A9), peptidoglycan recognition protein (PGLYRP1) and Ras homolog gene family, member B (RHOB). Specificity for AMPK was shown by testing inactive compound 3 in HanWistar rats. The exposure-effect relationship for GPNMB was investigated in a subchronic study in diabetic ZDF rats. GPNMB showed a dose-dependent up-regulation both acutely and after subchronic dosing. GPNMB up-regulation correlated with an increased Thr172-phosphorylation of AMPK in liver and quadriceps muscle in rats. CONCLUSION GPNMB has been identified as a translatable target engagement biomarker for use in clinical studies.
Collapse
Affiliation(s)
- Rolf Grempler
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
- * E-mail:
| | - Michael Wolff
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Eric Simon
- Department of Target Discovery Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Ramona Schmid
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Claudia Eisele
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Kathrin Rieber
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Elke Fischer
- Department of Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Sonja Mettel
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Ogsen Gabrielyan
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Denis Delic
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Gerd Luippold
- Department of Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Norbert Redeman
- Department of Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
17
|
Singh AM, Dalton S. What Can 'Brown-ing' Do For You? Trends Endocrinol Metab 2018; 29:349-359. [PMID: 29606342 PMCID: PMC5937921 DOI: 10.1016/j.tem.2018.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/27/2018] [Accepted: 03/01/2018] [Indexed: 12/31/2022]
Abstract
Human stem cell-based models of thermogenic adipocytes provide an opportunity for the establishment of new therapeutics, modeling of disease mechanisms, and understanding of development. Pluripotent stem cells, adipose-derived stem cells/preadipocytes, and programming-reprogramming-based approaches have been used to develop cell-based platforms for drug screening and transplantable therapeutics in the metabolic disease arena. Here we provide a detailed overview of these approaches, the latest advances in this field, and the opportunities and shortcomings they present. Moreover, we comment on how stem-cell-based platforms can be best utilized in the future for the treatment and understanding of metabolic diseases, including type 2 diabetes and associated medical issues such as obesity.
Collapse
Affiliation(s)
- Amar M Singh
- Center for Molecular Medicine, Department of Biochemistry and Molecular Biology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Stephen Dalton
- Center for Molecular Medicine, Department of Biochemistry and Molecular Biology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA.
| |
Collapse
|
18
|
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
|