1
|
Zhang X, Tian L, Majumdar A, Scheller EL. Function and Regulation of Bone Marrow Adipose Tissue in Health and Disease: State of the Field and Clinical Considerations. Compr Physiol 2024; 14:5521-5579. [PMID: 39109972 PMCID: PMC11725182 DOI: 10.1002/cphy.c230016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Bone marrow adipose tissue (BMAT) is a metabolically and clinically relevant fat depot that exists within bone. Two subtypes of BMAT, regulated and constitutive, reside in hematopoietic-rich red marrow and fatty yellow marrow, respectively, and exhibit distinct characteristics compared to peripheral fat such as white and brown adipose tissues. Bone marrow adipocytes (BMAds) are evolutionally preserved in most vertebrates, start development after birth and expand throughout life, and originate from unique progenitor populations that control bone formation and hematopoiesis. Mature BMAds also interact closely with other cellular components of the bone marrow niche, serving as a nearby energy reservoir to support the skeletal system, a signaling hub that contributes to both local and systemic homeostasis, and a final fuel reserve for survival during starvation. Though BMAT and bone are often inversely correlated, more BMAT does not always mean less bone, and the prevention of BMAT expansion as a strategy to prevent bone loss remains questionable. BMAT adipogenesis and lipid metabolism are regulated by the nervous systems and a variety of circulating hormones. This contributes to the plasticity of BMAT, including BMAT expansion in common physiological or pathological conditions, and BMAT catabolism under certain extreme circumstances, which are often associated with malnutrition and/or systemic inflammation. Altogether, this article provides a comprehensive overview of the local and systemic functions of BMAT and discusses the regulation and plasticity of this unique adipose tissue depot in health and disease. © 2024 American Physiological Society. Compr Physiol 14:5521-5579, 2024.
Collapse
Affiliation(s)
- Xiao Zhang
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Linda Tian
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Anurag Majumdar
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
2
|
Bian H, Shao X, Cai W, Fu H. Understanding the Reversible Binding of a Multichain Protein-Protein Complex through Free-Energy Calculations. J Phys Chem B 2024; 128:3598-3604. [PMID: 38574232 DOI: 10.1021/acs.jpcb.4c00519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
We demonstrate that the binding affinity of a multichain protein-protein complex, insulin dimer, can be accurately predicted using a streamlined route of standard binding free-energy calculations. We find that chains A and C, which do not interact directly during binding, stabilize the insulin monomer structures and reduce the binding affinity of the two monomers, therefore enabling their reversible association. Notably, we confirm that although classical methods can estimate the binding affinity of the insulin dimer, conventional molecular dynamics, enhanced sampling algorithms, and classical geometrical routes of binding free-energy calculations may not fully capture certain aspects of the role played by the noninteracting chains in the binding dynamics. Therefore, this study not only elucidates the role of noninteracting chains in the reversible binding of the insulin dimer but also offers a methodological guide for investigating the reversible binding of multichain protein-protein complexes utilizing streamlined free-energy calculations.
Collapse
Affiliation(s)
- Hengwei Bian
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Xueguang Shao
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| | - Wensheng Cai
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| | - Haohao Fu
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| |
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
|
Yadav R, Swetanshu, Singh P. The molecular mechanism of obesity: The science behind natural exercise yoga and healthy diets in the treatment of obesity. Curr Probl Cardiol 2024; 49:102345. [PMID: 38103823 DOI: 10.1016/j.cpcardiol.2023.102345] [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: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
The review centers on the scientific evidence underlying obesity, providing a detailed examination of the role of perilipin in this condition. It explores potential causes of obesity and delves into therapeutic approaches involving exercise, yoga, and herbal treatments. The paper discusses natural sources that can contribute to combating obesity and underscores the importance of exercise in a scientific context for overcoming obesity. Additionally, it includes information on herbal ingredients that aid in reducing obesity. The review also examines the impact of exercise type and intensity at various time intervals on muscle development. It elucidates triglyceride hydrolysis through different enzymes and the deposition of fatty acids in adipose tissue. The mechanisms by which alpha/beta hydrolase domain-containing protein 5 (ABHD5) and hormone-sensitive lipase (HSL) target and activate their functions are detailed. The inflammatory response in obesity is explored, encompassing inflammatory markers, lipid storage diseases, and their classification with molecular mechanisms. Furthermore, the hormonal regulation of lipolysis is elaborated upon in the review.
Collapse
Affiliation(s)
- Rajesh Yadav
- Sharda School of Allied Health Sciences, Sharda University, Greater Noida-201310, Uttar Pradesh, India; Department of Physiology, All India Institute of Medical Science, New Delhi, India
| | - Swetanshu
- Department of Zoology, Banaras Hindu University, U.P, India
| | - Pratichi Singh
- School of Biological and Life Sciences, Galgotias University, Greater Noida-203201, Uttar Pradesh, India.
| |
Collapse
|
5
|
de Morais H, Bôas SKFV, de Souza CO, Miksza DR, Moreira CCL, Kurauti MA, Silva FDF, Cassolla P, Silva FGD, Limiere LC, Grassiolli S, Bazotte RB, de Souza HM. Peripheral insulin resistance is early, progressive, and correlated with cachexia in Walker-256 tumor-bearing rats. Cell Biochem Funct 2023; 41:1252-1262. [PMID: 37787620 DOI: 10.1002/cbf.3859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/14/2023] [Accepted: 09/06/2023] [Indexed: 10/04/2023]
Abstract
Insulin (INS) resistance is often found in cancer-bearing, but its correlation with cachexia development is not completely established. This study investigated the temporal sequence of the development of INS resistance and cachexia to establish the relationship between these factors in Walker-256 tumor-bearing rats (TB rats). INS hepatic sensitivity and INS resistance-inducing factors, such as free fatty acids (FFA) and tumor necrosis factor-α (TNF-α), were also evaluated. Studies were carried out on Days 2, 5, 8, and/or 12 after inoculation of tumor cells in rats. The peripheral INS sensitivity was assessed by the INS tolerance test and the INS hepatic sensitivity in in situ liver perfusion. TB rats with 5, 8, and 12 days of tumor, but not 2 days, showed decreased peripheral INS sensitivity (INS resistance), retroperitoneal fat, and body weight, compared to healthy rats, which were more pronounced on Day 12. Gastrocnemius muscle wasting was observed only on Day 12 of tumor. The peripheral INS resistance was significantly correlated (r = -.81) with weight loss. Liver INS sensitivity of TB rats with 2 and 5 days of tumor was unchanged, compared to healthy rats. TB rats with 12 days of tumor showed increased plasma FFA and increased TNF-α in retroperitoneal fat and liver, but not in the gastrocnemius, compared to healthy rats. In conclusion, peripheral INS resistance is early, starts along with fat and weight loss and before muscle wasting, progressive, and correlated with cachexia, suggesting that it may play an important role in the pathogenesis of the cachectic process in TB rats. Therefore, early correction of INS resistance may be a therapeutic approach to prevent and treat cancer cachexia.
Collapse
Affiliation(s)
- Hely de Morais
- Department of Physiological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | | | - Camila O de Souza
- Department of Physiological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Daniele Romani Miksza
- Department of Physiological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Carolina C L Moreira
- Department of Physiological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Mirian Ayumi Kurauti
- Department of Physiological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Flaviane de F Silva
- Department of Physiological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Priscila Cassolla
- Department of Physiological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | | | | | - Sabrina Grassiolli
- Department of Physiological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Roberto B Bazotte
- Department of Physiological Sciences, State University of Maringa, Maringá, Paraná, Brazil
| | - Helenir M de Souza
- Department of Physiological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| |
Collapse
|
6
|
Munasinghe PE, Saw EL, Reily-Bell M, Tonkin D, Kakinuma Y, Fronius M, Katare R. Non-neuronal cholinergic system delays cardiac remodelling in type 1 diabetes. Heliyon 2023; 9:e17434. [PMID: 37426799 PMCID: PMC10329120 DOI: 10.1016/j.heliyon.2023.e17434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 07/11/2023] Open
Abstract
AIMS Type 1 diabetes mellitus (T1DM) is associated with increased risk of cardiovascular disease (CVD) and mortality. The underlying mechanisms for T1DM-induced heart disease still remains unclear. In this study, we aimed to investigate the effects of cardiac non-neuronal cholinergic system (cNNCS) activation on T1DM-induced cardiac remodelling. METHODS T1DM was induced in C57Bl6 mice using low-dose streptozotocin. Western blot analysis was used to measure the expression of cNNCS components at different time points (4, 8, 12, and 16 weeks after T1DM induction). To assess the potential benefits of cNNCS activation, T1DM was induced in mice with cardiomyocyte-specific overexpression of choline acetyltransferase (ChAT), the enzyme required for acetylcholine (Ac) synthesis. We evaluated the effects of ChAT overexpression on cNNCS components, vascular and cardiac remodelling, and cardiac function. KEY FINDINGS Western blot analysis revealed dysregulation of cNNCS components in hearts of T1DM mice. Intracardiac ACh levels were also reduced in T1DM. Activation of ChAT significantly increased intracardiac ACh levels and prevented diabetes-induced dysregulation of cNNCS components. This was associated with preserved microvessel density, reduced apoptosis and fibrosis, and improved cardiac function. SIGNIFICANCE Our study suggests that cNNCS dysregulation may contribute to T1DM-induced cardiac remodelling, and that increasing ACh levels may be a potential therapeutic strategy to prevent or delay T1DM-induced heart disease.
Collapse
Affiliation(s)
- Pujika Emani Munasinghe
- Department of Physiology, HeartOtago, University of Otago, 270, Great King Street, Dunedin, 9010, New Zealand
| | - Eng Leng Saw
- Department of Physiology, HeartOtago, University of Otago, 270, Great King Street, Dunedin, 9010, New Zealand
| | - Matthew Reily-Bell
- Department of Physiology, HeartOtago, University of Otago, 270, Great King Street, Dunedin, 9010, New Zealand
| | - Devin Tonkin
- Department of Physiology, HeartOtago, University of Otago, 270, Great King Street, Dunedin, 9010, New Zealand
| | - Yoshihiko Kakinuma
- Department of Bioregulatory Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Martin Fronius
- Department of Bioregulatory Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Rajesh Katare
- Department of Physiology, HeartOtago, University of Otago, 270, Great King Street, Dunedin, 9010, New Zealand
| |
Collapse
|
7
|
Abe T, Sato T, Murotomi K. Sudachitin and Nobiletin Stimulate Lipolysis via Activation of the cAMP/PKA/HSL Pathway in 3T3-L1 Adipocytes. Foods 2023; 12:foods12101947. [PMID: 37238764 DOI: 10.3390/foods12101947] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Polymethoxyflavones are flavonoids that are abundant in citrus fruit peels and have beneficial effects on human health. Previous studies have demonstrated that the polymethoxyflavones, namely sudachitin and nobiletin, ameliorate obesity and diabetes in humans and rodents. Although nobiletin induces lipolysis in adipocytes, lipolytic pathway activation by sudachitin has not been clarified in adipocytes. In this study, the effect of sudachitin on lipolysis was elucidated in murine 3T3-L1 adipocytes. Glycerol release into the medium and activation of the cyclic AMP (cAMP)/protein kinase A (PKA)/hormone-sensitive lipase (HSL) pathway was evaluated in 3T3-L1-differentiated adipocytes. Treatment with sudachitin and nobiletin for 24 and 48 h did not induce cytotoxicity at concentrations of up to 50 μM. Sudachitin and nobiletin at concentrations of 30 and 50 μM increased intracellular cAMP and medium glycerol levels in 3T3-L1 adipocytes. Western blotting revealed that sudachitin and nobiletin dose-dependently increased protein levels of phosphorylated PKA substrates and phosphorylated HSL. Sudachitin- and nobiletin-induced glycerol release, phosphorylation of PKA substrates, and HSL phosphorylation were suppressed by pharmacological inhibition of adenylate cyclase and PKA. These findings indicated that sudachitin, similar to nobiletin, exerts anti-obesogenic effects, at least in part through the induction of lipolysis in adipocytes.
Collapse
Affiliation(s)
- Tomoki Abe
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki 305-8566, Japan
| | - Tomoyuki Sato
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki 305-8566, Japan
| | - Kazutoshi Murotomi
- Molecular Neurophysiology Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki 305-8566, Japan
| |
Collapse
|
8
|
Bridge-Comer PE, Reilly SM. Measuring the Rate of Lipolysis in Ex vivo Murine Adipose Tissue and Primary Preadipocytes Differentiated In Vitro. J Vis Exp 2023:10.3791/65106. [PMID: 37010285 PMCID: PMC10583296 DOI: 10.3791/65106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
Abstract
Adipocytes store energy in the form of triglycerides in lipid droplets. This energy can be mobilized via lipolysis, where the fatty acid side chains are sequentially cleaved from the glycerol backbone, resulting in the release of free fatty acids and glycerol. Due to the low expression of glycerol kinase in white adipocytes, glycerol re-uptake rates are negligible, while fatty acid re-uptake is dictated by the fatty acid binding capacity of media components such as albumin. Both glycerol and fatty acid release into media can be quantified by colorimetric assays to determine the lipolytic rate. By measuring these factors at multiple time points, one can determine the linear rate of lipolysis with high confidence. Here, we provide a detailed protocol for the measurement of lipolysis in in vitro differentiated adipocytes and ex vivo adipose tissue from mice. This protocol may also be optimized for other preadipocyte cell lines or adipose tissue from other organisms; considerations and optimization parameters are discussed. This protocol is designed to be useful in determining and comparing the rate of adipocyte lipolysis between mouse models and treatments.
Collapse
Affiliation(s)
- Pania E Bridge-Comer
- Weill Center for Metabolic Health, Department of Medicine, Weill Cornell Medicine
| | - Shannon M Reilly
- Weill Center for Metabolic Health, Department of Medicine, Weill Cornell Medicine;
| |
Collapse
|
9
|
Azimian L, Weerasuriya NM, Munasinghe R, Song S, Lin CY, You L. Investigating the effects of Ceylon cinnamon water extract on HepG2 cells for Type 2 diabetes therapy. Cell Biochem Funct 2023; 41:254-267. [PMID: 36779418 DOI: 10.1002/cbf.3778] [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: 06/11/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 02/14/2023]
Abstract
Cinnamon and its extracts have been used as herbal remedies for many ailments, including for reducing insulin resistance and diabetes complications. Type 2 diabetes mellitus (T2DM) is a rapidly growing health concern around the world. Although many drugs are available for T2DM treatment, side effects and costs can be considerable, and there is increasing interest in natural products for managing chronic health conditions. Cinnamon may decrease the expression of genes associated with T2DM risk. The purpose of this study was to evaluate the effects of cinnamon water extract (CWE) compared with metformin on T2DM-related gene expression. HepG2 human hepatoma cells, widely used in drug metabolism and hepatotoxicity studies, were treated with different concentrations of metformin or CWE for 24 or 48 h. Cell viability was assessed by MTT (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assay and glucose uptake was compared in untreated and CWE- or metformin-treated cells under high-glucose conditions. Finally, total RNA was extracted and analyzed by RNA sequencing (RNA-seq), and bioinformatics analyses were performed to compare the transcriptional effects of CWE and metformin. We found cell viability was better in cells treated with CWE than in metformin-treated cells, demonstrating that CWE was not toxic at tested doses. CWE significantly increased glucose uptake in HepG2 cells, to the same degree as metformin (1.4-fold). RNA-seq data revealed CWE and metformin both induced significantly increased (1.3- to 1.4-fold) glucose uptake gene expression compared with untreated controls. Transcriptional differences between CWE and metformin were not significant. The effects of 0.125 mg mL-1 CWE on gene expression were comparable to 1.5 mg mL-1 (9.5 mM) metformin. In addition, gene expression at 0.125 mg mL-1 CWE was comparable to 1.5 mg mL-1 (9.5 mM) metformin. Our results reveal that CWE's effects on cell viability, glucose uptake, and gene expression in HepG2 cells are comparable to those of metformin, suggesting CWE may be an effective dietary supplement for mitigating T2DM-related metabolic dysfunction.
Collapse
Affiliation(s)
- Leila Azimian
- Department of Mechanical and Industrial Engineering, The University of Toronto, Toronto, Ontario, Canada
| | | | | | - Suzie Song
- Department of Mechanical and Industrial Engineering, The University of Toronto, Toronto, Ontario, Canada
| | - Chun-Yu Lin
- Institute of Biomedical Engineering, The University of Toronto, Toronto, Ontario, Canada
| | - Lidan You
- Department of Mechanical and Industrial Engineering, The University of Toronto, Toronto, Ontario, Canada.,Institute of Biomedical Engineering, The University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Kersten S. The impact of fasting on adipose tissue metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159262. [PMID: 36521736 DOI: 10.1016/j.bbalip.2022.159262] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/20/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Fasting and starvation were common occurrences during human evolution and accordingly have been an important environmental factor shaping human energy metabolism. Humans can tolerate fasting reasonably well through adaptative and well-orchestrated time-dependent changes in energy metabolism. Key features of the adaptive response to fasting are the breakdown of liver glycogen and muscle protein to produce glucose for the brain, as well as the gradual depletion of the fat stores, resulting in the release of glycerol and fatty acids into the bloodstream and the production of ketone bodies in the liver. In this paper, an overview is presented of our current understanding of the effects of fasting on adipose tissue metabolism. Fasting leads to reduced uptake of circulating triacylglycerols by adipocytes through inhibition of the activity of the rate-limiting enzyme lipoprotein lipase. In addition, fasting stimulates the degradation of stored triacylglycerols by activating the key enzyme adipose triglyceride lipase. The mechanisms underlying these events are discussed, with a special interest in insights gained from studies on humans. Furthermore, an overview is presented of the effects of fasting on other metabolic pathways in the adipose tissue, including fatty acid synthesis, glucose uptake, glyceroneogenesis, autophagy, and the endocrine function of adipose tissue.
Collapse
Affiliation(s)
- Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, the Netherlands.
| |
Collapse
|
11
|
Arya P, Kumar P. Diosgenin: An ingress towards solving puzzle for diabetes treatment. J Food Biochem 2022; 46:e14390. [PMID: 36106684 DOI: 10.1111/jfbc.14390] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/16/2022] [Accepted: 08/26/2022] [Indexed: 01/13/2023]
Abstract
The consumption and composition of food in daily life predict our health in long run. The relation of diabetes to sweets is quite popular. Diabetes hampers the glucose and insulin regulation in the human body by damaging pancreatic β cells. Diabetes has a strong potential towards altering cellular mechanisms of organs causing unlawful performance. Diabetes alters pathways like TLR4, AChE, NF-ĸB, LPL, and PPAR at different sites that affect the normal cellular machinery and cause damage to the local tissue and organ. The long-lasting effect of diabetes was observed in vascular, cardia, nervous, skeletal, reproductive, hepatic, ocular, and renal systems. The increasing awareness of diabetes and its concern has awakened the common people more enthusiastically. Due to rising harm from diabetes, scientific researchers tend to have more eyes toward it. While searching for diabetes solutions, fenugreek diosgenin could pop up with some positive effects in curing the same. Diosgenin helps to lower the scathe of diabetes by modifying cellular pathways in favor of healthy bodily functions. Diosgenin altered the pathways for renewal of pancreatic β cells for better insulin secretion, initiate GLUT4, enhanced DHEA, modify ER-α-mediated PI3K/Akt pathways. Diosgenin can be an appropriate insult for diabetes in a much evolving way for a healthy lifestyle. PRACTICAL APPLICATIONS: Diabetes is one of the most death causing diseases in the medical world. Regrettably the cure of diabetes is yet to be found. Various scientific team working on the same to look after the most appropriate way for diabetes treatment. There is enormous growth of nutraceutical in the market claiming for cure of different metabolic disorders. Among various bioactive compound fenugreek's diosgenin could took a leap over other in curing and preventing the damage caused by diabetes to different organs. The role of diosgenin in curing various metabolic disorders is quite popular from some time. This article also emphasizes over beneficiary effect of diosgenin in curing the damages caused by diabetes by altering cellular metabolism processes. Hence diosgenin could be a better way for researchers to develop a method for diabetes treatment.
Collapse
Affiliation(s)
- Prajya Arya
- Department of Food Engineering and Technology, Sant Longowal Institute of Engineering and Technology, Longowal, India
| | - Pradyuman Kumar
- Department of Food Engineering and Technology, Sant Longowal Institute of Engineering and Technology, Longowal, India
| |
Collapse
|
12
|
Meriin AB, Zaarur N, Roy D, Kandror KV. Egr1 plays a major role in the transcriptional response of white adipocytes to insulin and environmental cues. Front Cell Dev Biol 2022; 10:1003030. [PMID: 36246998 PMCID: PMC9554007 DOI: 10.3389/fcell.2022.1003030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
It is believed that insulin regulates metabolic functions of white adipose tissue primarily at the post-translational level via the PI3K-Akt-mediated pathway. Still, changes in transcription also play an important role in the response of white adipocytes to insulin and environmental signals. One transcription factor that is dramatically and rapidly induced in adipocytes by insulin and nutrients is called Early Growth Response 1, or Egr1. Among other functions, it directly binds to promoters of leptin and ATGL stimulating the former and inhibiting the latter. Furthermore, expression of Egr1 in adipocytes demonstrates cell autonomous circadian pattern suggesting that Egr1 not only mediates the effect of insulin and nutrients on lipolysis and leptin production but also, coordinates insulin action with endogenous circadian rhythms of adipose tissue.
Collapse
Affiliation(s)
- A. B. Meriin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - N. Zaarur
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - D. Roy
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - K. V. Kandror
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
- *Correspondence: K. V. Kandror,
| |
Collapse
|
13
|
Chen H, Pei Q, Tao L, Xia J, Lu G, Zong Y, Xie W, Li W, Huang C, Zeng T, Yu X, Wang W, Chen G, Yang S, Cheng R, Li X. ASC Regulates Subcutaneous Adipose Tissue Lipogenesis and Lipolysis via p53/AMPKα Axis. Int J Mol Sci 2022; 23:ijms231710042. [PMID: 36077447 PMCID: PMC9456541 DOI: 10.3390/ijms231710042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Obesity has become an extensive threat to human health due to associated chronic inflammation and metabolic diseases. Apoptosis-associated speck-like protein (ASC) is a critical link between inflammasome and apoptosis-inducing proteins. In this study, we aimed to clarify the role of ASC in lipid metabolism. With high-fat diet (HFD) and knockout leptin gene mice (ob/ob), we found that ASC expression in subcutaneous adipose tissue (SAT) correlated with obesity. It could also positively regulate the reprogramming of cellular energy metabolism. Stromal vascular fractions (SVF) cells derived from the SAT of Asc-/- mice or SVF from wild-type (WT) mice transfected with ASC siRNA were used to further investigate the underlying molecular mechanisms. We found ASC deficiency could lead to lipogenesis and inhibit lipolysis in SAT, aggravating lipid accumulation and impairing metabolic balance. In addition, our results showed that p53 and AMPKα expression were inhibited in SAT when ASC level was low. p53 and AMP-activated protein kinase α (AMPKα) were then assessed to elucidate whether they were downstream of ASC in regulating lipid metabolism. Our results revealed that ASC deficiency could promote lipid accumulation by increasing lipogenesis and decreasing lipolysis through p53/AMPKα axis. Regulation of ASC on lipid metabolism might be a novel therapeutic target for obesity.
Collapse
Affiliation(s)
- Hong Chen
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qilin Pei
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Linfen Tao
- Department of Laboratory Medicine, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350001, China
| | - Jing Xia
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Guocai Lu
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China
| | - Ying Zong
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China
| | - Wenhua Xie
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Wanqing Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Chenglong Huang
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ting Zeng
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xinyu Yu
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Weixuan Wang
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Gaojun Chen
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Song Yang
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Rui Cheng
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
- Correspondence: (R.C.); (X.L.); Tel.: +86-23-6848-1525 (R.C.); +86-23-6848-5589 (X.L.)
| | - Xi Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
- Correspondence: (R.C.); (X.L.); Tel.: +86-23-6848-1525 (R.C.); +86-23-6848-5589 (X.L.)
| |
Collapse
|
14
|
Kabir MT, Ferdous Mitu J, Akter R, Akhtar MF, Saleem A, Al-Harrasi A, Bhatia S, Rahman MS, Damiri F, Berrada M, Rahman MH. Therapeutic potential of dopamine agonists in the treatment of type 2 diabetes mellitus. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:46385-46404. [PMID: 35486279 DOI: 10.1007/s11356-022-20445-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/21/2022] [Indexed: 06/14/2023]
Abstract
Diabetes is a global health concern that has affected almost 415 million people globally. Bromocriptine is a dopamine D2 agonist, which is a Food and Drug Administration (FDA)-approved drug to treat type 2 diabetes mellitus (T2DM) patients. However, it is considered that a novel treatment therapy is required which can be used in the treatment of diabetes with or without other antidiabetic agents. Dopamine agonists are usually used in neurological disorders like Parkinson's disease (PD), restless leg syndrome, and hyperprolactinemia. However, dopamine agonists including bromocriptine and cabergoline are also effective in reducing the glycemic level in T2DM patients. Bromocriptine was formerly used for the treatment of PD, hyperprolactinemia, and restless leg syndrome, but now it is used for improving glycemic levels as well as reducing free fatty acids and triglycerides. In addition, cabergoline has been found to be effective in glycemic control, but this drug is yet to be approved by the FDA due to its limitations and lack of study. Findings of the clinical trials of bromocriptine have suggested that it reduces almost 0.4-0.8% glycated hemoglobin and cardiovascular risk by 40% in insulin-resistant patients. Moreover, the safe use of bromocriptine in obese T2DM patients makes it a more attractive option as it causes weight loss. Indeed, bromocriptine is a novel therapy for T2DM patients, as its mechanism of action is unique in T2DM patients with minimal adverse effects. This review summarizes the potential of dopamine agonists in the treatment of T2DM.
Collapse
Affiliation(s)
- Md Tanvir Kabir
- Department of Pharmacy, Brac University, 66 Mohakhali, Dhaka, 1212, Bangladesh
| | | | - Raushanara Akter
- Department of Pharmacy, Brac University, 66 Mohakhali, Dhaka, 1212, Bangladesh
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Lahore Campus, Lahore, Pakistan
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, 616 Birkat Al Mauz, P.O. Box 33, Nizwa, Oman
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Center, University of Nizwa, 616 Birkat Al Mauz, P.O. Box 33, Nizwa, Oman
- School of Health Science, University of Petroleum and Energy Studies, Prem Nagar, Dehradun, Uttarakhand, 248007, India
| | - Md Sohanur Rahman
- Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal, 8200, Bangladesh
| | - Fouad Damiri
- Laboratory of Biomolecules and Organic Synthesis (BIOSYNTHO), Department of Chemistry, Faculty of Sciences Ben M'Sick, University Hassan II of Casablanca, Casablanca, Morocco
| | - Mohammed Berrada
- Laboratory of Biomolecules and Organic Synthesis (BIOSYNTHO), Department of Chemistry, Faculty of Sciences Ben M'Sick, University Hassan II of Casablanca, Casablanca, Morocco
| | - Md Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka, 1213, Bangladesh.
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, 26426, Gangwon-do, Korea.
| |
Collapse
|
15
|
John CM, Arockiasamy S. Sinapic acid prevents adipogenesis by regulating transcription factors and exerts an anti-ROS effect by modifying the intracellular anti-oxidant system in 3T3-L1 adipocytes. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:611-620. [PMID: 35911638 PMCID: PMC9282747 DOI: 10.22038/ijbms.2022.62590.13847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 05/16/2022] [Indexed: 11/21/2022]
Abstract
Objectives In this study, we tested the hypothesis that sinapic acid (SA), a naturally occurring hydroxycinnamic acid found in vegetables, cereal grains, and oilseed crops with various biological activities suppresses adipogenesis in 3T3-L1 adipocytes by down-regulating adipogenesis transcription factor. Materials and Methods 3T3-L1 adipocytes were treated with SA and evaluated by Oil Red O staining, triglyceride estimation, lipolysis, and reverse transcription-polymerase chain reaction. 3T3-L1 adipocytes were treated with various concentrations of SA (100 to 1000 μmol) during differentiation. Results SA prevented an increase in adipocytes by reducing preadipocyte clonal expansion. ORO staining analyses revealed that SA reduced cytoplasmic lipid droplet accumulation in 3T3-L1 by 57% at the highest concentration of 1000 μmol without affecting cell viability. Furthermore, SA down-regulated the expression of peroxisome proliferator-activated receptor-gamma, CCAAT/enhancer-binding protein alpha, sterol regulatory element-binding protein 1c, and fatty acid synthase. ROS generated during adipogenesis was also attenuated by SA treatment by increasing antioxidant enzymes superoxide dismutase, catalase, and the cellular antioxidant glutathione. SA demonstrated no in vivo toxicity in the Drosophila melanogaster model. Conclusion These results suggest that SA exerts anti-oxidant and anti-adipogenic effects and could be used as a functional nutraceutical ingredient in combatting obesity-related diseases.
Collapse
Affiliation(s)
- Cordelia Mano John
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai – 600116, Tamil Nadu, India
| | - Sumathy Arockiasamy
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai – 600116, Tamil Nadu, India,Corresponding author: Sumathy Arockiasamy. Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology Sri Ramachandra Institute of Higher Education and Research Porur, Chennai – 600116 Tamil Nadu. Tel: 044 – 24768027/29; Extn:8760;
| |
Collapse
|
16
|
Sancar G, Liu S, Gasser E, Alvarez JG, Moutos C, Kim K, van Zutphen T, Wang Y, Huddy TF, Ross B, Dai Y, Zepeda D, Collins B, Tilley E, Kolar MJ, Yu RT, Atkins AR, van Dijk TH, Saghatelian A, Jonker JW, Downes M, Evans RM. FGF1 and insulin control lipolysis by convergent pathways. Cell Metab 2022; 34:171-183.e6. [PMID: 34986332 PMCID: PMC8863067 DOI: 10.1016/j.cmet.2021.12.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/08/2021] [Accepted: 12/06/2021] [Indexed: 01/07/2023]
Abstract
Inexorable increases in insulin resistance, lipolysis, and hepatic glucose production (HGP) are hallmarks of type 2 diabetes. Previously, we showed that peripheral delivery of exogenous fibroblast growth factor 1 (FGF1) has robust anti-diabetic effects mediated by the adipose FGF receptor (FGFR) 1. However, its mechanism of action is not known. Here, we report that FGF1 acutely lowers HGP by suppressing adipose lipolysis. On a molecular level, FGF1 inhibits the cAMP-protein kinase A axis by activating phosphodiesterase 4D (PDE4D), which separates it mechanistically from the inhibitory actions of insulin via PDE3B. We identify Ser44 as an FGF1-induced regulatory phosphorylation site in PDE4D that is modulated by the feed-fast cycle. These findings establish the FGF1/PDE4 pathway as an alternate regulator of the adipose-HGP axis and identify FGF1 as an unrecognized regulator of fatty acid homeostasis.
Collapse
Affiliation(s)
- Gencer Sancar
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sihao Liu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Emanuel Gasser
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jacqueline G Alvarez
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Christopher Moutos
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Kyeongkyu Kim
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Tim van Zutphen
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, the Netherlands
| | - Yuhao Wang
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Timothy F Huddy
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Brittany Ross
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Yang Dai
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - David Zepeda
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Brett Collins
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Emma Tilley
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Matthew J Kolar
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ruth T Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Annette R Atkins
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Theo H van Dijk
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, the Netherlands
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Johan W Jonker
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, the Netherlands
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
17
|
PI3K and AKT at the Interface of Signaling and Metabolism. Curr Top Microbiol Immunol 2022; 436:311-336. [DOI: 10.1007/978-3-031-06566-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
18
|
Attie AD, Tang QQ, Bornfeldt KE. The insulin centennial-100 years of milestones in biochemistry. J Biol Chem 2021; 297:101278. [PMID: 34717954 PMCID: PMC8605089 DOI: 10.1016/j.jbc.2021.101278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/21/2022] Open
|
19
|
Attie AD, Tang QQ, Bornfeldt KE. The insulin centennial-100 years of milestones in biochemistry. J Lipid Res 2021; 62:100132. [PMID: 34717951 PMCID: PMC8721491 DOI: 10.1016/j.jlr.2021.100132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/05/2022] Open
|
20
|
Abstract
The molecular mechanisms of cellular insulin action have been the focus of much investigation since the discovery of the hormone 100 years ago. Insulin action is impaired in metabolic syndrome, a condition known as insulin resistance. The actions of the hormone are initiated by binding to its receptor on the surface of target cells. The receptor is an α2β2 heterodimer that binds to insulin with high affinity, resulting in the activation of its tyrosine kinase activity. Once activated, the receptor can phosphorylate a number of intracellular substrates that initiate discrete signaling pathways. The tyrosine phosphorylation of some substrates activates phosphatidylinositol-3-kinase (PI3K), which produces polyphosphoinositides that interact with protein kinases, leading to activation of the kinase Akt. Phosphorylation of Shc leads to activation of the Ras/MAP kinase pathway. Phosphorylation of SH2B2 and of Cbl initiates activation of G proteins such as TC10. Activation of Akt and other protein kinases produces phosphorylation of a variety of substrates, including transcription factors, GTPase-activating proteins, and other kinases that control key metabolic events. Among the cellular processes controlled by insulin are vesicle trafficking, activities of metabolic enzymes, transcriptional factors, and degradation of insulin itself. Together these complex processes are coordinated to ensure glucose homeostasis.
Collapse
|
21
|
Genomic and Non-Genomic Actions of Glucocorticoids on Adipose Tissue Lipid Metabolism. Int J Mol Sci 2021; 22:ijms22168503. [PMID: 34445209 PMCID: PMC8395154 DOI: 10.3390/ijms22168503] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/01/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Glucocorticoids (GCs) are hormones that aid the body under stress by regulating glucose and free fatty acids. GCs maintain energy homeostasis in multiple tissues, including those in the liver and skeletal muscle, white adipose tissue (WAT), and brown adipose tissue (BAT). WAT stores energy as triglycerides, while BAT uses fatty acids for heat generation. The multiple genomic and non-genomic pathways in GC signaling vary with exposure duration, location (adipose tissue depot), and species. Genomic effects occur directly through the cytosolic GC receptor (GR), regulating the expression of proteins related to lipid metabolism, such as ATGL and HSL. Non-genomic effects act through mechanisms often independent of the cytosolic GR and happen shortly after GC exposure. Studying the effects of GCs on adipose tissue breakdown and generation (lipolysis and adipogenesis) leads to insights for treatment of adipose-related diseases, such as obesity, coronary disease, and cancer, but has led to controversy among researchers, largely due to the complexity of the process. This paper reviews the recent literature on the genomic and non-genomic effects of GCs on WAT and BAT lipolysis and proposes research to address the many gaps in knowledge related to GC activity and its effects on disease.
Collapse
|
22
|
Ng XW, Chung YH, Piston DW. Intercellular Communication in the Islet of Langerhans in Health and Disease. Compr Physiol 2021; 11:2191-2225. [PMID: 34190340 PMCID: PMC8985231 DOI: 10.1002/cphy.c200026] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Blood glucose homeostasis requires proper function of pancreatic islets, which secrete insulin, glucagon, and somatostatin from the β-, α-, and δ-cells, respectively. Each islet cell type is equipped with intrinsic mechanisms for glucose sensing and secretory actions, but these intrinsic mechanisms alone cannot explain the observed secretory profiles from intact islets. Regulation of secretion involves interconnected mechanisms among and between islet cell types. Islet cells lose their normal functional signatures and secretory behaviors upon dispersal as compared to intact islets and in vivo. In dispersed islet cells, the glucose response of insulin secretion is attenuated from that seen from whole islets, coordinated oscillations in membrane potential and intracellular Ca2+ activity, as well as the two-phase insulin secretion profile, are missing, and glucagon secretion displays higher basal secretion profile and a reverse glucose-dependent response from that of intact islets. These observations highlight the critical roles of intercellular communication within the pancreatic islet, and how these communication pathways are crucial for proper hormonal and nonhormonal secretion and glucose homeostasis. Further, misregulated secretions of islet secretory products that arise from defective intercellular islet communication are implicated in diabetes. Intercellular communication within the islet environment comprises multiple mechanisms, including electrical synapses from gap junctional coupling, paracrine interactions among neighboring cells, and direct cell-to-cell contacts in the form of juxtacrine signaling. In this article, we describe the various mechanisms that contribute to proper islet function for each islet cell type and how intercellular islet communications are coordinated among the same and different islet cell types. © 2021 American Physiological Society. Compr Physiol 11:2191-2225, 2021.
Collapse
Affiliation(s)
- Xue W Ng
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| | - Yong H Chung
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| | - David W Piston
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| |
Collapse
|
23
|
Effect of eugenol treatment in hyperglycemic murine models: A meta-analysis. Pharmacol Res 2021; 165:105315. [PMID: 33497803 DOI: 10.1016/j.phrs.2020.105315] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/23/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022]
Abstract
Diabetes is a highly prevalent health condition affecting many people worldwide. In vitro studies have described the positive effects of cloves and its major compound, eugenol, in the treatment of diabetes. However, it is unclear whether the effects of this compound are negative, neutral, or positive, on hyperglycemic animals. Therefore, a meta-analytical review was conducted to determine the magnitude of effects of eugenol on variables directly and indirectly related to diabetes. This study revealed that eugenol treatment decreased the glucose levels and the activity of carbohydrate-metabolizing enzymes, ameliorated the lipid profile, and reduced the oxidative, renal, and hepatic damages in hyperglycemic rodents. Moreover, eugenol alleviated the weight loss and restored the activity of the antioxidant defense system. Insulin levels was not affected by eugenol treatment. Also, mixed model analyses revealed that the use of purified or non-purified eugenol and the concentrations administered significantly affected the treatment outcome. In conclusion, our findings indicate that eugenol may have potential therapeutic effects in the treatment of diabetes. Furthermore, this study can direct future preclinical and clinical trials, with important implications for human health.
Collapse
|
24
|
Adipose PTEN acts as a downstream mediator of a brain-fat axis in environmental enrichment. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2020; 4. [PMID: 35355831 PMCID: PMC8963210 DOI: 10.1016/j.cpnec.2020.100013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background/Objectives Environmental enrichment (EE) is a physiological model to investigate brain-fat interactions. We previously discovered that EE activates the hypothalamic-sympathoneural adipocyte (HSA) axis via induction of brain-derived neurotrophic factor (BDNF), thus leading to sympathetic stimulation of white adipose tissue (WAT) and an anti-obesity phenotype. Here, we investigate whether PTEN acts as a downstream mediator of the HSA axis in the EE. Methods Mice were housed in EE for 4- and 16-week periods to determine how EE regulates adipose PTEN. Hypothalamic injections of adeno-associated viral (AAV) vectors expressing BDNF and a dominant negative form of its receptor were performed to assess the role of the HSA axis in adipose PTEN upregulation. A β-blocker, propranolol, and a denervation agent, 6-hydroydopamine, were administered to assess sympathetic signaling in the observed EE-PTEN phenotype. To determine whether inducing PTEN is sufficient to reproduce certain EE adipose remodeling, we overexpressed PTEN in WAT using an AAV vector. To determine whether adipose PTEN is necessary for the EE-mediated reduction in adipocyte size, we injected a rAAV vector expressing Cre recombinase to the WAT of adult PTENflox mice and placed the mice in EE. Results EE upregulated adipose PTEN expression, which was associated with suppression of AKT and ERK phosphorylation, increased hormone-sensitive lipase (HSL) phosphorylation, and reduced adiposity. PTEN regulation was found to be controlled by the HSA axis—with the hypothalamic BDNF acting as the upstream mediator—and dependent on sympathetic innervation. AAV-mediated adipose PTEN overexpression recapitulated EE-mediated adipose changes including suppression of AKT and ERK phosphorylation, increased HSL phosphorylation, and reduced adipose mass, whereas PTEN knockdown blocked the EE-induced reduction of adipocyte size. Conclusions These data suggest that adipose PTEN responds to environmental stimuli and serves as downstream mediator of WAT remodeling in the EE paradigm, resulting in decreased adipose mass and decreased adipocyte size. Environmental enrichment (EE) induces adipose PTEN expression and is associated with (1) suppression of AKT phosphorylation, (2) increased hormone-sensitive lipase phosphorylation, and (3) decreased adiposity The hypothalamic-sympathoneural-adipocyte (HSA) axis mediates EE-induced adipose PTEN rAAV-mediated gene delivery of PTEN to adipose tissues mimics EE-related adipose remodeling Knockdown of adipose PTEN blocks EE-induced reductions in adipocyte size
Collapse
|
25
|
Sanna M, Borgo C, Compagnin C, Favaretto F, Vindigni V, Trento M, Bettini S, Comin A, Belligoli A, Rugge M, Bassetto F, Donella-Deana A, Vettor R, Busetto L, Milan G. White Adipose Tissue Expansion in Multiple Symmetric Lipomatosis Is Associated with Upregulation of CK2, AKT and ERK1/2. Int J Mol Sci 2020; 21:ijms21217933. [PMID: 33114687 PMCID: PMC7662313 DOI: 10.3390/ijms21217933] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/09/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Multiple symmetric lipomatosis (MSL) is a rare disorder characterized by overgrowing lipomatous tissue (LT) in the subcutaneous adipose tissue (SAT). What LT is and how it expands are not completely understood; previous data suggested that it could derive from brown AT precursors. In six MSL type I patients, we compared LT morphology by histological and immunohistochemistry (IHC) analysis, gene expression, by qPCR, kinase activity, by Western Blot and in vitro assay to paired-control SAT using AT from patients with pheochromocytoma as a human browning reference. In the stromal vascular fraction (SVF), we quantified adipose stem cells (ASCs) by flow cytometry, the proliferation rate, white and beige adipogenic potential and clonogenicity and adipogenicity by a limiting dilution assay. LT displayed white AT morphology and expression pattern and did not show increased levels of the brown-specific marker UCP1. In LT, we evidenced AKT, CK2 and ERK1/2 hyperactivation. LT-SVF contained increased ASCs, proliferated faster, sprouted clones and differentiated into adipocytes better than the control, displaying enhanced white adipogenic potential but not increased browning compared to SAT. In conclusion, LT is a white AT depot expanding by hyperplasia through increased stemness and enhanced white adipogenesis upregulating AKT, CK2 and ERK1/2, which could represent new targets to counteract MSL.
Collapse
Affiliation(s)
- Marta Sanna
- Endocrine-Metabolic Laboratory, Department of Medicine, Internal Medicine 3, University of Padua, 35128 Padua, Italy; (M.S.); (C.C.); (F.F.); (S.B.); (A.C.); (A.B.); (R.V.); (L.B.)
| | - Christian Borgo
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (C.B.); (A.D.-D.)
| | - Chiara Compagnin
- Endocrine-Metabolic Laboratory, Department of Medicine, Internal Medicine 3, University of Padua, 35128 Padua, Italy; (M.S.); (C.C.); (F.F.); (S.B.); (A.C.); (A.B.); (R.V.); (L.B.)
| | - Francesca Favaretto
- Endocrine-Metabolic Laboratory, Department of Medicine, Internal Medicine 3, University of Padua, 35128 Padua, Italy; (M.S.); (C.C.); (F.F.); (S.B.); (A.C.); (A.B.); (R.V.); (L.B.)
| | - Vincenzo Vindigni
- Clinic of Plastic and Reconstructive Surgery, Department of Neurosciences, University of Padua, 35128 Padua, Italy; (V.V.); (F.B.)
| | - Mariangela Trento
- Surgical Pathology and Cytopathology Unit, Department of Medicine, University of Padua, 35121 Padua, Italy; (M.T.); (M.R.)
| | - Silvia Bettini
- Endocrine-Metabolic Laboratory, Department of Medicine, Internal Medicine 3, University of Padua, 35128 Padua, Italy; (M.S.); (C.C.); (F.F.); (S.B.); (A.C.); (A.B.); (R.V.); (L.B.)
| | - Alessandra Comin
- Endocrine-Metabolic Laboratory, Department of Medicine, Internal Medicine 3, University of Padua, 35128 Padua, Italy; (M.S.); (C.C.); (F.F.); (S.B.); (A.C.); (A.B.); (R.V.); (L.B.)
| | - Anna Belligoli
- Endocrine-Metabolic Laboratory, Department of Medicine, Internal Medicine 3, University of Padua, 35128 Padua, Italy; (M.S.); (C.C.); (F.F.); (S.B.); (A.C.); (A.B.); (R.V.); (L.B.)
| | - Massimo Rugge
- Surgical Pathology and Cytopathology Unit, Department of Medicine, University of Padua, 35121 Padua, Italy; (M.T.); (M.R.)
| | - Franco Bassetto
- Clinic of Plastic and Reconstructive Surgery, Department of Neurosciences, University of Padua, 35128 Padua, Italy; (V.V.); (F.B.)
| | - Arianna Donella-Deana
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (C.B.); (A.D.-D.)
| | - Roberto Vettor
- Endocrine-Metabolic Laboratory, Department of Medicine, Internal Medicine 3, University of Padua, 35128 Padua, Italy; (M.S.); (C.C.); (F.F.); (S.B.); (A.C.); (A.B.); (R.V.); (L.B.)
| | - Luca Busetto
- Endocrine-Metabolic Laboratory, Department of Medicine, Internal Medicine 3, University of Padua, 35128 Padua, Italy; (M.S.); (C.C.); (F.F.); (S.B.); (A.C.); (A.B.); (R.V.); (L.B.)
| | - Gabriella Milan
- Endocrine-Metabolic Laboratory, Department of Medicine, Internal Medicine 3, University of Padua, 35128 Padua, Italy; (M.S.); (C.C.); (F.F.); (S.B.); (A.C.); (A.B.); (R.V.); (L.B.)
- Correspondence: ; Tel.: +39-049-8218550; Fax: +39-049-8218555
| |
Collapse
|
26
|
Adipocyte lipolysis: from molecular mechanisms of regulation to disease and therapeutics. Biochem J 2020; 477:985-1008. [PMID: 32168372 DOI: 10.1042/bcj20190468] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/19/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022]
Abstract
Fatty acids (FAs) are stored safely in the form of triacylglycerol (TAG) in lipid droplet (LD) organelles by professional storage cells called adipocytes. These lipids are mobilized during adipocyte lipolysis, the fundamental process of hydrolyzing TAG to FAs for internal or systemic energy use. Our understanding of adipocyte lipolysis has greatly increased over the past 50 years from a basic enzymatic process to a dynamic regulatory one, involving the assembly and disassembly of protein complexes on the surface of LDs. These dynamic interactions are regulated by hormonal signals such as catecholamines and insulin which have opposing effects on lipolysis. Upon stimulation, patatin-like phospholipase domain containing 2 (PNPLA2)/adipocyte triglyceride lipase (ATGL), the rate limiting enzyme for TAG hydrolysis, is activated by the interaction with its co-activator, alpha/beta hydrolase domain-containing protein 5 (ABHD5), which is normally bound to perilipin 1 (PLIN1). Recently identified negative regulators of lipolysis include G0/G1 switch gene 2 (G0S2) and PNPLA3 which interact with PNPLA2 and ABHD5, respectively. This review focuses on the dynamic protein-protein interactions involved in lipolysis and discusses some of the emerging concepts in the control of lipolysis that include allosteric regulation and protein turnover. Furthermore, recent research demonstrates that many of the proteins involved in adipocyte lipolysis are multifunctional enzymes and that lipolysis can mediate homeostatic metabolic signals at both the cellular and whole-body level to promote inter-organ communication. Finally, adipocyte lipolysis is involved in various diseases such as cancer, type 2 diabetes and fatty liver disease, and targeting adipocyte lipolysis is of therapeutic interest.
Collapse
|
27
|
Inazumi T, Yamada K, Shirata N, Sato H, Taketomi Y, Morita K, Hohjoh H, Tsuchiya S, Oniki K, Watanabe T, Sasaki Y, Oike Y, Ogata Y, Saruwatari J, Murakami M, Sugimoto Y. Prostaglandin E2-EP4 Axis Promotes Lipolysis and Fibrosis in Adipose Tissue Leading to Ectopic Fat Deposition and Insulin Resistance. Cell Rep 2020; 33:108265. [DOI: 10.1016/j.celrep.2020.108265] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 09/10/2020] [Accepted: 09/22/2020] [Indexed: 12/24/2022] Open
|
28
|
Wasner HK. Prostaglandylinositol cyclic phosphate, the natural antagonist of cyclic AMP. IUBMB Life 2020; 72:2282-2289. [PMID: 32893983 DOI: 10.1002/iub.2372] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 11/06/2022]
Abstract
While searching for a counterpart to cyclic AMP, a new compound was found to inhibit adenylate cyclase. It was identified as prostaglandyl-(15-4')-myo-inositol (1':2'-cyclic)-phosphate (cyclic PIP). The substrates for its biosynthesis are prostaglandin E (PGE) and the novel inositol phosphate, guanosine diphospho-4-myo-inositol 1:2-cyclic phosphate (n-IP). The basic regulatory properties of cyclic PIP are to inhibit dose-dependently protein kinase A (PKA) and to seven-fold activate protein ser/thr phosphatase holoenzyme. These regulations occur as rapidly as the activation of PKA by cyclic AMP. Such regulatory properties are essential for the meticulous regulation of the equilibrium between the phospho- and de-phospho-form of interconvertible enzymes. The synthesis of cyclic PIP is stimulated by insulin and noradrenaline (α-receptor action). The insulin-stimulated cyclic PIP synthase is active in a tyrosine-phosphorylated state. A comparable characterization of the adrenaline-stimulated cyclic PIP synthase is still incomplete. In streptozotocin-diabetic rats, the hormonal stimulation of cyclic PIP synthesis decreases with time. Cyclic PIP synthesis is activated by biguanides as metformin two to four-fold and by antihypertensive drugs two-fold. Inhibition of cyclic PIP synthesis leads to a metabolic state as observed in early-stage type-2 diabetes. In summary, all living cells synthesize cyclic PIP, which switches on anabolism, whereas cyclic AMP triggers catabolism.
Collapse
Affiliation(s)
- Heinrich K Wasner
- BioReg Biopharm, HTI (Health, Technology, Innovation), University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
29
|
Kopietz F, Rupar K, Berggreen C, Säll J, Vertommen D, Degerman E, Rider MH, Göransson O. Inhibition of AMPK activity in response to insulin in adipocytes: involvement of AMPK pS485, PDEs, and cellular energy levels. Am J Physiol Endocrinol Metab 2020; 319:E459-E471. [PMID: 32663099 DOI: 10.1152/ajpendo.00065.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Insulin resistance in obesity and type 2 diabetes has been shown to be associated with decreased de novo fatty acid (FA) synthesis in adipose tissue. It is known that insulin can acutely stimulate FA synthesis in adipocytes; however, the mechanisms underlying this effect are unclear. The rate-limiting step in FA synthesis is catalyzed by acetyl-CoA carboxylase (ACC), known to be regulated through inhibitory phosphorylation at S79 by the AMP-activated protein kinase (AMPK). Previous results from our laboratory showed an inhibition of AMPK activity by insulin, which was accompanied by PKB-dependent phosphorylation of AMPK at S485. However, whether the S485 phosphorylation is required for insulin-induced inhibition of AMPK or other mechanisms underlie the reduced kinase activity is not known. To investigate this, primary rat adipocytes were transduced with a recombinant adenovirus encoding AMPK-WT or a nonphosphorylatable AMPK S485A mutant. AMPK activity measurements by Western blot analysis and in vitro kinase assay revealed that WT and S485A AMPK were inhibited to a similar degree by insulin, indicating that AMPK S485 phosphorylation is not required for insulin-induced AMPK inhibition. Further analysis suggested an involvement of decreased AMP-to-ATP ratios in the insulin-induced inhibition of AMPK activity, whereas a possible contribution of phosphodiesterases was excluded. Furthermore, we show that insulin-induced AMPK S485 phosphorylation also occurs in human adipocytes, suggesting it to be of an importance yet to be revealed. Altogether, this study increases our understanding of how insulin regulates AMPK activity, and with that, FA synthesis, in adipose tissue.
Collapse
Affiliation(s)
| | - Kaja Rupar
- Department of Experimental Medical Science, Lund University, Sweden
| | | | - Johanna Säll
- Department of Experimental Medical Science, Lund University, Sweden
| | - Didier Vertommen
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Eva Degerman
- Department of Experimental Medical Science, Lund University, Sweden
| | - Mark H Rider
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Olga Göransson
- Department of Experimental Medical Science, Lund University, Sweden
| |
Collapse
|
30
|
Insulin and β-adrenergic receptors mediate lipolytic and anti-lipolytic signalling that is not altered by type 2 diabetes in human adipocytes. Biochem J 2020; 476:2883-2908. [PMID: 31519735 PMCID: PMC6792037 DOI: 10.1042/bcj20190594] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 12/21/2022]
Abstract
Control of fatty acid storage and release in adipose tissue is fundamental in energy homeostasis and the development of obesity and type 2 diabetes. We here take the whole signalling network into account to identify how insulin and β-adrenergic stimulation in concert controls lipolysis in mature subcutaneous adipocytes obtained from non-diabetic and, in parallel, type 2 diabetic women. We report that, and show how, the anti-lipolytic effect of insulin can be fully explained by protein kinase B (PKB/Akt)-dependent activation of the phosphodiesterase PDE3B. Through the same PKB-dependent pathway β-adrenergic receptor signalling, via cAMP and PI3Kα, is anti-lipolytic and inhibits its own stimulation of lipolysis by 50%. Through this pathway both insulin and β-adrenergic signalling control phosphorylation of FOXO1. The dose–response of lipolysis is bell-shaped, such that insulin is anti-lipolytic at low concentrations, but at higher concentrations of insulin lipolysis was increasingly restored due to inhibition of PDE3B. The control of lipolysis was not altered in adipocytes from diabetic individuals. However, the release of fatty acids was increased by 50% in diabetes due to reduced reesterification of lipolytically liberated fatty acids. In conclusion, our results reveal mechanisms of control by insulin and β-adrenergic stimulation — in human adipocytes — that define a network of checks and balances ensuring robust control to secure uninterrupted supply of fatty acids without reaching concentrations that put cellular integrity at risk. Moreover, our results define how selective insulin resistance leave lipolytic control by insulin unaltered in diabetes, while the fatty acid release is substantially increased.
Collapse
|
31
|
Hosooka T, Hosokawa Y, Matsugi K, Shinohara M, Senga Y, Tamori Y, Aoki C, Matsui S, Sasaki T, Kitamura T, Kuroda M, Sakaue H, Nomura K, Yoshino K, Nabatame Y, Itoh Y, Yamaguchi K, Hayashi Y, Nakae J, Accili D, Yokomizo T, Seino S, Kasuga M, Ogawa W. The PDK1-FoxO1 signaling in adipocytes controls systemic insulin sensitivity through the 5-lipoxygenase-leukotriene B 4 axis. Proc Natl Acad Sci U S A 2020; 117:11674-11684. [PMID: 32393635 PMCID: PMC7261087 DOI: 10.1073/pnas.1921015117] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Although adipocytes are major targets of insulin, the influence of impaired insulin action in adipocytes on metabolic homeostasis remains unclear. We here show that adipocyte-specific PDK1 (3'-phosphoinositide-dependent kinase 1)-deficient (A-PDK1KO) mice manifest impaired metabolic actions of insulin in adipose tissue and reduction of adipose tissue mass. A-PDK1KO mice developed insulin resistance, glucose intolerance, and hepatic steatosis, and this phenotype was suppressed by additional ablation of FoxO1 specifically in adipocytes (A-PDK1/FoxO1KO mice) without an effect on adipose tissue mass. Neither circulating levels of adiponectin and leptin nor inflammatory markers in adipose tissue differed between A-PDK1KO and A-PDK1/FoxO1KO mice. Lipidomics and microarray analyses revealed that leukotriene B4 (LTB4) levels in plasma and in adipose tissue as well as the expression of 5-lipoxygenase (5-LO) in adipose tissue were increased and restored in A-PDK1KO mice and A-PDK1/FoxO1KO mice, respectively. Genetic deletion of the LTB4 receptor BLT1 as well as pharmacological intervention to 5-LO or BLT1 ameliorated insulin resistance in A-PDK1KO mice. Furthermore, insulin was found to inhibit LTB4 production through down-regulation of 5-LO expression via the PDK1-FoxO1 pathway in isolated adipocytes. Our results indicate that insulin signaling in adipocytes negatively regulates the production of LTB4 via the PDK1-FoxO1 pathway and thereby maintains systemic insulin sensitivity.
Collapse
Affiliation(s)
- Tetsuya Hosooka
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
| | - Yusei Hosokawa
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
| | - Kaku Matsugi
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
| | - Masakazu Shinohara
- Division of Epidemiology, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
| | - Yoko Senga
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
| | - Yoshikazu Tamori
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
- Department of Internal Medicine, Chibune General Hospital, 555-0001 Osaka, Japan
| | - Chikako Aoki
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
| | - Sho Matsui
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, 371-8512 Maebashi, Japan
| | - Tsutomu Sasaki
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, 371-8512 Maebashi, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, 371-8512 Maebashi, Japan
| | - Masashi Kuroda
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, 770-8503 Tokushima, Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, 770-8503 Tokushima, Japan
| | - Kazuhiro Nomura
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
| | - Kei Yoshino
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
| | - Yuko Nabatame
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
| | - Yoshito Itoh
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 602-8566 Kyoto, Japan
| | - Kanji Yamaguchi
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 602-8566 Kyoto, Japan
| | - Yoshitake Hayashi
- Division of Molecular Medicine and Medical Genetics, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
| | - Jun Nakae
- Department of Internal Medicine, Keio University School of Medicine, 160-8582 Tokyo, Japan
| | - Domenico Accili
- Department of Medicine and Naomi Berrie Diabetes Center, Columbia University, NY 10032
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University School of Medicine, 113-8421 Tokyo, Japan
| | - Susumu Seino
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan
| | - Masato Kasuga
- The Institute for Adult Diseases, Asahi Life Foundation, 103-0002 Tokyo, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, 650-0017 Kobe, Japan;
| |
Collapse
|
32
|
Islam H, Smith MMW, Scribbans TD, McCrady E, Castellani LN, Allen MD, Wright DC, Simpson CA, Gurd BJ. Effect of Acute High-intensity Interval Exercise on Whole-body Fat Oxidation and Subcutaneous Adipose Tissue Cell Signaling in Overweight Women. INTERNATIONAL JOURNAL OF EXERCISE SCIENCE 2020; 13:554-566. [PMID: 32509113 PMCID: PMC7241622 DOI: 10.70252/fqef4828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Exercise-induced alterations in adipose tissue insulin and/or β-adrenergic signaling may contribute to increases in whole-body fat oxidation following acute exercise. Thus, we examined changes in insulin (Akt, AS160) and β-adrenergic (PKA) signaling proteins in subcutaneous adipose tissue and whole-body fat oxidation in overweight women following acute high-intensity interval exercise (HIIE). Overweight females completed two experimental sessions in a randomized order: 1) control (bed rest) and 2) HIIE (10 × 4 min running intervals at 90% HRmax, 2-min recovery). Subcutaneous abdominal adipose tissue biopsies were obtained from 10 participants before (pre-), immediately (0hr) after (post-), 2hr post-, and 4hr post-exercise. Plasma glucose and insulin levels were assessed in venous blood samples obtained at each biopsy time-point from a different group of 5 participants (BMI-matched to biopsy group). Fat oxidation rates were estimated using the respiratory exchange ratio (RER) in all participants using indirect calorimetry pre-, 2hr post-, and 4hr post-exercise. RER was decreased (p < 0.05) at 2hr post-exercise after HIIE (0.77 ± 0.04) compared to control (0.84 ± 0.04). Despite higher plasma glucose (p < 0.01) and insulin (p < 0.05) levels at 0hr post-exercise versus control, no significant interaction effects were observed for Akt or AS160 phosphorylation (p > 0.05). Phosphorylation of PKA substrates was unaltered in both conditions (p > 0.05). Collectively, altered β-adrenergic and insulin signaling in subcutaneous adnominal adipose tissue does not appear to explain increased whole-body fat oxidation following acute HIIE.
Collapse
Affiliation(s)
- Hashim Islam
- School of Kinesiology of Health Studies, Queen's University, Kingston, ONTARIO, CANADA
| | - Marysa M W Smith
- School of Kinesiology of Health Studies, Queen's University, Kingston, ONTARIO, CANADA
| | - Trisha D Scribbans
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, MANITOBA, CANADA
| | - Emma McCrady
- School of Kinesiology of Health Studies, Queen's University, Kingston, ONTARIO, CANADA
| | - Laura N Castellani
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ONTARIO, CANADA
| | - Matti D Allen
- School of Kinesiology of Health Studies, Queen's University, Kingston, ONTARIO, CANADA
- School of Medicine, Queen's University, Kingston, ONTARIO, CANADA
| | - David C Wright
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ONTARIO, CANADA
| | - Craig A Simpson
- School of Kinesiology of Health Studies, Queen's University, Kingston, ONTARIO, CANADA
| | - Brendon J Gurd
- School of Kinesiology of Health Studies, Queen's University, Kingston, ONTARIO, CANADA
| |
Collapse
|
33
|
Getz M, Rangamani P, Ghosh P. Regulating cellular cyclic adenosine monophosphate: "Sources," "sinks," and now, "tunable valves". WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1490. [PMID: 32323924 DOI: 10.1002/wsbm.1490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 01/31/2020] [Accepted: 03/31/2020] [Indexed: 12/16/2022]
Abstract
A number of hormones and growth factors stimulate target cells via the second messenger pathways, which in turn regulate cellular phenotypes. Cyclic adenosine monophosphate (cAMP) is a ubiquitous second messenger that facilitates numerous signal transduction pathways; its production in cells is tightly balanced by ligand-stimulated receptors that activate adenylate cyclases (ACs), that is, "source" and by phosphodiesterases (PDEs) that hydrolyze it, that is, "sinks." Because it regulates various cellular functions, including cell growth and differentiation, gene transcription and protein expression, the cAMP signaling pathway has been exploited for the treatment of numerous human diseases. Reduction in cAMP is achieved by blocking "sources"; however, elevation in cAMP is achieved by either stimulating "source" or blocking "sinks." Here we discuss an alternative paradigm for the regulation of cellular cAMP via GIV/Girdin, the prototypical member of a family of modulators of trimeric GTPases, Guanine nucleotide Exchange Modulators (GEMs). Cells upregulate or downregulate cellular levels of GIV-GEM, which modulates cellular cAMP via spatiotemporal mechanisms distinct from the two most often targeted classes of cAMP modulators, "sources" and "sinks." A network-based compartmental model for the paradigm of GEM-facilitated cAMP signaling has recently revealed that GEMs such as GIV serve much like a "tunable valve" that cells may employ to finetune cellular levels of cAMP. Because dysregulated signaling via GIV and other GEMs has been implicated in multiple disease states, GEMs constitute a hitherto untapped class of targets that could be exploited for modulating aberrant cAMP signaling in disease states. This article is categorized under: Models of Systems Properties and Processes > Mechanistic Models Biological Mechanisms > Cell Signaling.
Collapse
Affiliation(s)
- Michael Getz
- Chemical Engineering Graduate Program, University of California San Diego, La Jolla, California, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA
| | - Pradipta Ghosh
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA
- Moores Comprehensive Cancer Center, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
34
|
Rago V, De Rose D, Santoro M, Panza S, Malivindi R, Andò S, D'Agata R, Aquila S. Human Sperm Express the Receptor for Glucagon-like Peptide-1 (GLP-1), Which Affects Sperm Function and Metabolism. Endocrinology 2020; 161:5802751. [PMID: 32157297 DOI: 10.1210/endocr/bqaa031] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022]
Abstract
AIM Glucagon-like peptide-1 (GLP-1) produces pleiotropic effects binding to the GLP-1 receptor (GLP1-R), potentiating insulin secretion in the pancreas. GLP1-R is expressed in peripheral tissues and evidence for its role in reproduction has come from knockout mice, although the relationship between GLP-1 and male fertility needs to be clarified. Given that human sperm is an insulin-sensitive and insulin-secreting cell, we hypothesized that the GLP-1/GLP1-R axis may be expressed and functional in these cells. RESULTS AND DISCUSSION We revealed the presence of GLP1-R by Western blotting and immunofluorescence analyses. Because Exendin-4 (Ex-4) displays similar functional properties to native GLP-1, we used this agonist to perform a dose-response study on progressive motility and cholesterol efflux, showing that 300 pM Ex-4 was the most effective treatment. These actions are mediated by GLP1-R and independent from sperm-secreted insulin. The exposure to Ex-4 fueled phosphatidylinositol-3-kinase (PI3K)/AKT signaling and was reversed by H89, indicating a protein kinase A (PKA)-dependence of GLP-1/GLP1-R signaling. It emerged that in sperm, insulin secretion regulated by Ex-4 did not occur in a strictly glucose-dependent manner. A stimulatory action of Ex-4/GLP1-R on lactate dehydrogenase and glucose-6-phosphate dehydrogenase (G6PDH) activities was observed. Ex-4/GLP1-R decreased triglycerides content concomitantly to enhanced lipase and acyl-coenzyme A (acyl-CoA) dehydrogenase activities, addressing a lipolytic effect. CONCLUSION Collectively, we discovered that human sperm is a new GLP1 incretin target, broadening our knowledge about the effects of the GLP1-R agonist in the male reproductive field. Further findings in humans should be conducted in the future to confirm it and to improve the translational aspect of this study.
Collapse
Affiliation(s)
- Vittoria Rago
- Department of Pharmacy and Sciences of Health and Nutrition, University of Calabria Cosenza, Italy
| | - Daniela De Rose
- Department of Pharmacy and Sciences of Health and Nutrition, University of Calabria Cosenza, Italy
- Centro Sanitario, University of Calabria Cosenza, Italy
| | - Marta Santoro
- Department of Pharmacy and Sciences of Health and Nutrition, University of Calabria Cosenza, Italy
- Centro Sanitario, University of Calabria Cosenza, Italy
| | - Salvatore Panza
- Department of Pharmacy and Sciences of Health and Nutrition, University of Calabria Cosenza, Italy
- Centro Sanitario, University of Calabria Cosenza, Italy
| | - Rocco Malivindi
- Department of Pharmacy and Sciences of Health and Nutrition, University of Calabria Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy and Sciences of Health and Nutrition, University of Calabria Cosenza, Italy
- Centro Sanitario, University of Calabria Cosenza, Italy
| | - Rosario D'Agata
- Department of Experimental and Clinical Medicine, University of Catania, Catania, Italy
| | - Saveria Aquila
- Department of Pharmacy and Sciences of Health and Nutrition, University of Calabria Cosenza, Italy
- Centro Sanitario, University of Calabria Cosenza, Italy
| |
Collapse
|
35
|
Zhang R, Xing B, Zhao J, Zhang X, Zhou L, Yang S, Wang Y, Yang F. Astragaloside IV relieves gestational diabetes mellitus in genetic mice through reducing hepatic gluconeogenesis. Can J Physiol Pharmacol 2020; 98:466-472. [PMID: 32160476 DOI: 10.1139/cjpp-2019-0548] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The glucose intolerance developed during pregnancy is called gestational diabetes mellitus (GDM). GDM has become a severe risk for the health of both mother and baby. Astragaloside IV (AS-IV) is the dominant active component in Astragalus membranaceus and has been reported to have anti-inflammation and immune-regulation function. We aimed to demonstrate the function of AS-IV in the therapy of GDM and the molecular mechanism in this process. C57BL/KsJ-Lepdb/+ female mice were used as the GDM model. The mRNA levels of relative genes in this research were detected by quantitative real-time PCR. The protein levels of relative genes were analyzed by Western blot. Serum lipid level was measured with an ILab Chemistry Analyzer 300 PLUS. Glucose, insulin, and lipid profile levels in the GDM mice model were decreased by AS-IV treatment. AS-IV downregulated the expression of inflammatory genes and upregulated the expressions of anti-oxidant genes in the GDM mice model. AS-IV treatment reduced cAMP accumulation in liver and reduced hepatic gluconeogenesis in GDM mice. This study demonstrated that AS-IV treatment has an effective therapeutic function of GDM in a mice model through the regulation of cAMP accumulation and hepatic gluconeogenesis.
Collapse
Affiliation(s)
- Ruixue Zhang
- Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou 061000, Hebei, China
| | - Baoheng Xing
- Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou 061000, Hebei, China
| | - Jianyong Zhao
- Cangzhou Hospital of Integrated TCM-WM·HEBEI, No. 31 Huanghe Road, Cangzhou 061000, Hebei, China
| | - Xuelei Zhang
- Cangzhou Hospital of Integrated TCM-WM·HEBEI, No. 31 Huanghe Road, Cangzhou 061000, Hebei, China
| | - Ling Zhou
- Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou 061000, Hebei, China
| | - Shuangyan Yang
- Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou 061000, Hebei, China
| | - Yong Wang
- Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou 061000, Hebei, China
| | - Fengzhen Yang
- Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou 061000, Hebei, China
| |
Collapse
|
36
|
Dibe HA, Townsend LK, McKie GL, Wright DC. Epinephrine responsiveness is reduced in livers from trained mice. Physiol Rep 2020; 8:e14370. [PMID: 32061187 PMCID: PMC7023888 DOI: 10.14814/phy2.14370] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/12/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
The liver is the primary metabolic organ involved in the endogenous production of glucose through glycogenolysis and gluconeogenesis. Hepatic glucose production (HGP) is increased via neural-hormonal mechanisms such as increases in catecholamines. To date, the effects of prior exercise training on the hepatic response to epinephrine have not been fully elucidated. To examine the role of epinephrine signaling on indices of HGP in trained mice, male C57BL/6 mice were either subjected to 12 days of voluntary wheel running or remained sedentary. Epinephrine, or vehicle control, was injected intraperitoneally on day 12 prior to sacrifice with blood glucose being measured 15 min postinjection. Epinephrine caused a larger glucose response in sedentary mice and this was paralleled by a greater reduction in liver glycogen in sedentary compared to trained mice. There was a main effect of epinephrine to increase the phosphorylation of protein kinase-A (p-PKA) substrates in the liver, which was driven by increases in the sedentary, but not trained, mice. Similarly, epinephrine-induced increases in the mRNA expression of hepatic adrenergic receptors (Adra1/2a, Adrb1), and glucose-6-phosphatase (G6pc) were greater in sedentary compared to trained mice. The mRNA expression of cAMP-degrading enzymes phosphodiesterase 3B and 4B (Pde3b, Pde4b) was greater in trained compared to sedentary mice. Taken together, our data suggest that prior exercise training reduces the liver's response to epinephrine. This could be beneficial in the context of training-induced glycogen sparing during exercise.
Collapse
Affiliation(s)
- Hana A Dibe
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Logan K Townsend
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Greg L McKie
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - David C Wright
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
37
|
Tozzi M, Hansen JB, Novak I. Pannexin-1 mediated ATP release in adipocytes is sensitive to glucose and insulin and modulates lipolysis and macrophage migration. Acta Physiol (Oxf) 2020; 228:e13360. [PMID: 31400255 DOI: 10.1111/apha.13360] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/29/2019] [Accepted: 08/07/2019] [Indexed: 12/15/2022]
Abstract
AIM Extracellular ATP signalling is involved in many physiological and pathophysiological processes in several tissues, including adipose tissue. Adipocytes have crucial functions in lipid and glucose metabolism and they express purinergic receptors. However, the sources of extracellular ATP in adipose tissue are not well characterized. In the present study, we investigated the mechanism and regulation of ATP release in white adipocytes, and evaluated the role of extracellular ATP as potential autocrine and paracrine signal. METHODS Online ATP release was monitored in C3H10T1/2 cells and freshly isolated murine adipocytes. The ATP release mechanism and its regulation were tested in cells exposed to adrenergic agonists, insulin, glucose load and pharmacological inhibitors. Cell metabolism was monitored using Seahorse respirometry and expression analysis of pannexin-1 was performed on pre- and mature adipocytes. The ATP signalling was evaluated in live cell imaging (Ca2+ , pore formation), glycerol release and its effect on macrophages was tested in co-culture and migration assays. RESULTS Here, we show that upon adrenergic stimulation white murine adipocytes release ATP through the pannexin-1 pore that is regulated by a cAMP-PKA-dependent pathway. The ATP release correlates with increased cell metabolism and is sensitive to glucose. Extracellular ATP induces Ca2+ signalling and lipolysis in adipocytes and promotes macrophage migration. Importantly, ATP release is markedly inhibited by insulin, which operates via the activation of phosphodiesterase 3. CONCLUSIONS Our findings reveal an insulin-pannexin-1-purinergic signalling crosstalk in adipose tissue and we propose that deregulation of this signalling may contribute to adipose tissue inflammation and type 2 diabetes.
Collapse
Affiliation(s)
- Marco Tozzi
- Section for Cell Biology and Physiology, Department of Biology University of Copenhagen Copenhagen Denmark
| | - Jacob B. Hansen
- Section for Cell Biology and Physiology, Department of Biology University of Copenhagen Copenhagen Denmark
| | - Ivana Novak
- Section for Cell Biology and Physiology, Department of Biology University of Copenhagen Copenhagen Denmark
| |
Collapse
|
38
|
Cyclic nucleotide phosphodiesterases: New targets in the metabolic syndrome? Pharmacol Ther 2020; 208:107475. [PMID: 31926200 DOI: 10.1016/j.pharmthera.2020.107475] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Metabolic diseases have a tremendous impact on human morbidity and mortality. Numerous targets regulating adenosine monophosphate kinase (AMPK) have been identified for treating the metabolic syndrome (MetS), and many compounds are being used or developed to increase AMPK activity. In parallel, the cyclic nucleotide phosphodiesterase families (PDEs) have emerged as new therapeutic targets in cardiovascular diseases, as well as in non-resolved pathologies. Since some PDE subfamilies inactivate cAMP into 5'-AMP, while the beneficial effects in MetS are related to 5'-AMP-dependent activation of AMPK, an analysis of the various controversial relationships between PDEs and AMPK in MetS appears interesting. The present review will describe the various PDE families, AMPK and molecular mechanisms in the MetS and discuss the PDEs/PDE modulators related to the tissues involved, thus supporting the discovery of original molecules and the design of new therapeutic approaches in MetS.
Collapse
|
39
|
Borah AK, Singh A, Yasmin R, Doley R, Mattaparthi VSK, Saha S. 1α, 25-dihydroxy Vitamin D3 containing fractions of Catharanthus roseus leaf aqueous extract inhibit preadipocyte differentiation and induce lipolysis in 3T3-L1 cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:338. [PMID: 31783835 PMCID: PMC6883588 DOI: 10.1186/s12906-019-2754-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 11/14/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND To investigate the potential of Catharanthus roseus leaf aqueous crude extract (CRACE) as a regulator of adipocyte development and function. METHODS 3T3-L1 adipogenesis model was used to investigate the effect of CRACE on adipogenesis. 3T3-L1 preadipocytes (for adipogenic differentiation) and mature 3T3-L1 adipocytes (for adipocyte function) were treated with non-toxic doses of CRACE. The outcomes were corroborated by intracellular lipid accumulation, expression of pro-and anti-adipogenic effector molecules. To investigate CRACE mediated lipolysis, cAMP accumulation, glycerol release and phosphorylation of key effector molecules were tested in treated mature adipocytes. Finally, the extract was fractionated to identify the active molecule/s in the extract. RESULTS CRACE significantly reduced adipocyte differentiation by modulating PPARγ expression. At early stage CRACE directly targeted Lipin1 expression and consequently impacted KLF7, subsequently expression of GATA2, CEBPα, SREBP1c were targeted, with PPARγ expression, particularly curtailed. While CRACE significantly reduced several lipogenic genes like FAS and GPD1 in mature adipocytes, concomitantly, it greatly increased lipolysis resulting in decreased lipid accumulation in mature adipocytes. The increase in lipolysis was due to decreased Akt activation, increased cAMP level, and PKA activity. The fractionation of CRACE allowed identification of two fractions with potent anti-adipogenic activity. Both the fractions contained 1α, 25-dihydroxy Vitamin D3 as major component. CONCLUSIONS 1α, 25-dihydroxy Vitamin D3 containing CRACE can be developed into an effective anti-obesity formulation that decreases adipogenesis and increases lipid catabolism.
Collapse
Affiliation(s)
- Anuj Kumar Borah
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, Assam 784028 India
| | - Archana Singh
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, Assam 784028 India
| | - Rafika Yasmin
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, Assam 784028 India
| | - Robin Doley
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, Assam 784028 India
| | | | - Sougata Saha
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209 India
| |
Collapse
|
40
|
Patel BM, Goyal RK. Liver and insulin resistance: New wine in old bottle!!! Eur J Pharmacol 2019; 862:172657. [DOI: 10.1016/j.ejphar.2019.172657] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 09/02/2019] [Accepted: 09/05/2019] [Indexed: 12/20/2022]
|
41
|
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
|
42
|
Therapeutic opportunities in colon cancer: Focus on phosphodiesterase inhibitors. Life Sci 2019; 230:150-161. [PMID: 31125564 DOI: 10.1016/j.lfs.2019.05.043] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 02/08/2023]
Abstract
Despite novel technologies, colon cancer remains undiagnosed and 25% of patients are diagnosed with metastatic colon cancer. Resistant to chemotherapeutic agents is one of the major problems associated with treating colon cancer which creates the need to develop novel agents targeting towards newer targets. A phosphodiesterase is a group of isoenzyme, which, hydrolyze cyclic nucleotides and thereby lowers intracellular levels of cAMP and cGMP leading to tumorigenic effects. Many in vitro and in vivo studies have confirmed increased PDE expression in different types of cancers including colon cancer. cAMP-specific PDE inhibitors increase intracellular cAMP that leads to activation of effector molecules-cAMP-dependent protein kinase A, exchange protein activated by cAMP and cAMP gated ion channels. These molecules regulate cellular responses and exert its anticancer role through different mechanisms including apoptosis, inhibition of angiogenesis, upregulating tumor suppressor genes and suppressing oncogenes. On the other hand, cGMP specific PDE inhibitors exhibit anticancer effects through cGMP dependent protein kinase and cGMP dependent cation channels. Elevation in cGMP works through activation of caspases, suppression of Wnt/b-catenin pathway and TCF transcription leading to inhibition of CDK and survivin. These studies point out towards the fact that PDE inhibition is associated with anti-proliferative, anti-apoptotic and anti-angiogenic pathways involved in its anticancer effects in colon cancer. Thus, inhibition of PDE enzymes can be used as a novel approach to treat colon cancer. This review will focus on cAMP and cGMP signaling pathways leading to tumorigenesis and the use of PDE inhibitors in colon cancer.
Collapse
|
43
|
Gong XM, Li YF, Luo J, Wang JQ, Wei J, Wang JQ, Xiao T, Xie C, Hong J, Ning G, Shi XJ, Li BL, Qi W, Song BL. Gpnmb secreted from liver promotes lipogenesis in white adipose tissue and aggravates obesity and insulin resistance. Nat Metab 2019; 1:570-583. [PMID: 32694855 DOI: 10.1038/s42255-019-0065-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/05/2019] [Indexed: 12/13/2022]
Abstract
Metabolism in mammals is regulated by complex interplay among different organs. Fatty acid synthesis is increased in white adipose tissue (WAT) when it is inhibited in the liver. Here we identify glycoprotein non-metastatic melanoma protein B (Gpnmb) as one liver-WAT cross-talk factor involved in lipogenesis. Inhibition of the hepatic sterol regulatory element-binding protein pathway leads to increased transcription of Gpnmb and promotes processing of the membrane protein to a secreted form. Gpnmb stimulates lipogenesis in WAT and exacerbates diet-induced obesity and insulin resistance. In humans, Gpnmb is tightly associated with body mass index and is a strong risk factor for obesity. Gpnmb inhibition by a neutralizing antibody or liver-specific knockdown improves metabolic parameters, including weight gain reduction and increased insulin sensitivity, probably by promoting the beiging of WAT. These results suggest that Gpnmb is a liver-secreted factor regulating lipogenesis in WAT, and that Gpnmb inhibition may provide a therapeutic strategy in obesity and diabetes.
Collapse
Affiliation(s)
- Xue-Min Gong
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yun-Feng Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Jie Luo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Ji-Qiu Wang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, State Key Laboratory of Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Wei
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Ju-Qiong Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Ting Xiao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Chang Xie
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jie Hong
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, State Key Laboratory of Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, State Key Laboratory of Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiong-Jie Shi
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Bo-Liang Li
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wei Qi
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China.
| |
Collapse
|
44
|
Yoshihara A, Luo Y, Ishido Y, Usukura K, Oda K, Sue M, Kawashima A, Hiroi N, Suzuki K. Inhibitory effects of methimazole and propylthiouracil on iodotyrosine deiodinase 1 in thyrocytes. Endocr J 2019; 66:349-357. [PMID: 30814441 DOI: 10.1507/endocrj.ej18-0380] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Methimazole (MMI) and propylthiouracil (PTU) are commonly used for the treatment of Graves' disease. They share similar inhibitory effects on thyroid hormone biosynthesis by interfering with thyroid peroxidase (TPO)-mediated oxidation and organification of iodine. However, their potential effects on other thyroid functional molecules have not been explored in depth. To identify novel effects of MMI and PTU, DNA microarray analysis, real-time PCR, Western blotting, immunofluorescence staining and confocal laser scanning microscopy were performed using FRTL-5 rat thyroid cells. DNA microarray analysis indicated that both MMI and PTU suppress iodotyrosine deiodinase 1 (Iyd, Dehal1) mRNA levels. Further studies revealed that Dehal1 mRNA levels was stimulated by TSH, insulin and serum, while it was suppressed by iodine and a follicular concentration of thyroglobulin. MMI and PTU significantly suppressed Dehal1 expression induced by TSH, insulin and serum. On the other hand, although MMI suppressed Dehal1 expression in the absence of TSH, PTU only weakly suppressed Dehal1 without TSH. These results suggest that PTU and MMI may use different mechanisms to regulate Dehal1 expression, and TSH may play essential and differential roles in mediating PTU and MMI signals in thyrocytes. The drugs also inhibited re-distribution of Dehal1 protein into newly formed lysosomes following thyroglobulin endocytosis. These findings imply complex and multifaceted regulation of Dehal1 in the thyroid and suggest that MMI and PTU modulate Dehal1 expression and distribution of the protein in thyrocytes to exert their effect.
Collapse
Affiliation(s)
- Aya Yoshihara
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
- Center for Medical Education, Faculty of Medicine, Toho University, Ota, Tokyo 143-8540, Japan
- Department of Internal Medicine, Tokyo Metropolitan Bokutoh Hospital, Sumida, Tokyo 130-8575, Japan
| | - Yuqian Luo
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
| | - Yuko Ishido
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
| | - Kensei Usukura
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
| | - Kenzaburo Oda
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
- Department of Internal Medicine, Tokyo Metropolitan Bokutoh Hospital, Sumida, Tokyo 130-8575, Japan
| | - Mariko Sue
- Department of Medicine III, Faculty of Medicine of the Technische Universität Dresden, Dresden 01307, Germany
| | - Akira Kawashima
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
| | - Naoki Hiroi
- Center for Medical Education, Faculty of Medicine, Toho University, Ota, Tokyo 143-8540, Japan
| | - Koichi Suzuki
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
| |
Collapse
|
45
|
Yin C, Liu WH, Liu Y, Wang L, Xiao Y. PID1 alters the antilipolytic action of insulin and increases lipolysis via inhibition of AKT/PKA pathway activation. PLoS One 2019; 14:e0214606. [PMID: 30990811 PMCID: PMC6467375 DOI: 10.1371/journal.pone.0214606] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/17/2019] [Indexed: 11/19/2022] Open
Abstract
Purpose The aim of this study was to investigate the effect of phosphotyrosine interaction domain containing 1 (PID1) on the insulin-induced activation of the AKT (protein kinase B)/protein kinase A (PKA)/hormone-sensitive lipase (HSL) pathway and lipolysis. Methods Sprague–Dawley rats were fed either chow or a high-fat diet (HFD). The levels of insulin, glycerol, free fatty acids (FFAs) and PID1 mRNA expression were measured in the 2 groups. Furthermore, we examined the role of PID1 in the regulation of the AKT/PKA/HSL cascade and lipolysis in the 3T3-L1 cell line. Results Adipose tissue from HFD rats exhibited elevated PID1 expression, which showed a positive correlation with insulin levels and lipolysis. In 3T3-L1 adipocytes, we found that the antilipolytic effect of insulin is mediated by AKT and that phosphorylated AKT results in the promotion of PDE3B expression, the dephosphorylation of PKA and HSL and the suppression of glycerol release. However, overexpression of PID1 and treatment with 1 μM isoproterenol and 100 nM insulin for 24 h resulted in an increased release of glycerol and a noticeable inhibition of AKT phosphorylation, PDE3B expression and the phosphorylation of PKA/HSL in 3T3-L1 cells. In contrast, knockdown of PID1 and treatment with the above reagents inhibited lipolysis and activated the phosphorylation of AKT, which resulted in the dephosphorylation of PKA and HSL. Conclusions Our findings indicate that PID1 in adipose tissue increases lipolysis by altering the antilipolytic action of insulin. This suggests that PID1 may represent a new therapeutic target to ameliorate adipocyte lipolysis and hence improve insulin sensitivity.
Collapse
Affiliation(s)
- Chunyan Yin
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shan Xi, People’s Republic of China
| | - Wei hua Liu
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shan Xi, People’s Republic of China
| | - Yuesheng Liu
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shan Xi, People’s Republic of China
| | - Li Wang
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shan Xi, People’s Republic of China
| | - Yanfeng Xiao
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shan Xi, People’s Republic of China
- * E-mail:
| |
Collapse
|
46
|
Bódis K, Roden M. Energy metabolism of white adipose tissue and insulin resistance in humans. Eur J Clin Invest 2018; 48:e13017. [PMID: 30107041 DOI: 10.1111/eci.13017] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/22/2018] [Accepted: 08/12/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Insulin resistance not only occurs in obesity, but also in lipodystrophy. Although adipose tissue mass affects metabolic fluxes and participates in interorgan crosstalk, the role of energy metabolism within white adipose tissue for insulin resistance is less clear. MATERIALS AND METHODS A Medline search identified in vivo studies in humans on energy and lipid metabolism in subcutaneous (SAT) and visceral adipose tissue (VAT). Studies in adipocyte cultures and transgenic animal models were included for the better understanding of the link between abnormal energy metabolism in adipose tissue and insulin resistance. RESULTS The current literature indicates that higher lipolysis and lower lipogenesis in VAT compared to SAT enhance portal delivery of lipid metabolites (glycerol and fatty acids) to the liver. Thus, the lower lipolysis and higher lipogenesis in SAT favour storage of excess lipids and allow for protection of insulin-sensitive tissues from lipotoxic effects. In insulin-resistant humans, enhanced lipolysis and impaired lipogenesis in adipose tissue lead to release of cytokines and lipid metabolites, ultimately promoting insulin resistance. Adipose tissue of insulin-resistant humans also displays lower expression of proteins involved in mitochondrial function. In turn, this leads to lower availability of mitochondria-derived energy sources for lipogenesis in adipose tissue. CONCLUSIONS Abnormal mitochondrial function in human white adipose tissue likely contributes to the secretion of lipid metabolites and lactate, which are linked to insulin resistance in peripheral tissues. However, the relevance of adipose tissue energy metabolism for the regulation of human insulin sensitivity remains to be further elucidated.
Collapse
Affiliation(s)
- Kálmán Bódis
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Michael Roden
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
47
|
Xu J, Jia YF, Tapadar S, Weaver JD, Raji IO, Pithadia DJ, Javeed N, García AJ, Choi DS, Matveyenko AV, Oyelere AK, Shin CH. Inhibition of TBK1/IKKε Promotes Regeneration of Pancreatic β-cells. Sci Rep 2018; 8:15587. [PMID: 30349097 PMCID: PMC6197228 DOI: 10.1038/s41598-018-33875-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/01/2018] [Indexed: 12/18/2022] Open
Abstract
β-cell proliferation induction is a promising therapeutic strategy to restore β-cell mass. By screening small molecules in a transgenic zebrafish model of type 1 diabetes, we identified inhibitors of non-canonical IκB kinases (IKKs), TANK-binding kinase 1 (TBK1) and IκB kinase ε (IKKε), as enhancers of β-cell regeneration. The most potent β-cell regeneration enhancer was a cinnamic acid derivative (E)-3-(3-phenylbenzo[c]isoxazol-5-yl)acrylic acid (PIAA), which, acting through the cAMP-dependent protein kinase A (PKA), stimulated β-cell-specific proliferation by increasing cyclic AMP (cAMP) levels and mechanistic target of rapamycin (mTOR) activity. A combination of PIAA and cilostamide, an inhibitor of β-cell-enriched cAMP hydrolyzing enzyme phosphodiesterase (PDE) 3, enhanced β-cell proliferation, whereas overexpression of PDE3 blunted the mitogenic effect of PIAA in zebrafish. PIAA augmented proliferation of INS-1β-cells and β-cells in mammalian islets including human islets with elevation in cAMP levels and insulin secretion. PIAA improved glycemic control in streptozotocin (STZ)-induced diabetic mice with increases in β-cell proliferation, β-cell area, and insulin content in the pancreas. Collectively, these data reveal an evolutionarily conserved and critical role of TBK1/IKKε suppression in expanding functional β-cell mass.
Collapse
Affiliation(s)
- Jin Xu
- School of Biological Sciences and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Yun-Fang Jia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Subhasish Tapadar
- School of Chemistry and Biochemistry and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jessica D Weaver
- Woodruff School of Mechanical Engineering and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Idris O Raji
- School of Chemistry and Biochemistry and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Deeti J Pithadia
- School of Biological Sciences and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Naureen Javeed
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Chong Hyun Shin
- School of Biological Sciences and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA. .,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
48
|
Petersen MC, Shulman GI. Mechanisms of Insulin Action and Insulin Resistance. Physiol Rev 2018; 98:2133-2223. [PMID: 30067154 PMCID: PMC6170977 DOI: 10.1152/physrev.00063.2017] [Citation(s) in RCA: 1667] [Impact Index Per Article: 238.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 12/15/2022] Open
Abstract
The 1921 discovery of insulin was a Big Bang from which a vast and expanding universe of research into insulin action and resistance has issued. In the intervening century, some discoveries have matured, coalescing into solid and fertile ground for clinical application; others remain incompletely investigated and scientifically controversial. Here, we attempt to synthesize this work to guide further mechanistic investigation and to inform the development of novel therapies for type 2 diabetes (T2D). The rational development of such therapies necessitates detailed knowledge of one of the key pathophysiological processes involved in T2D: insulin resistance. Understanding insulin resistance, in turn, requires knowledge of normal insulin action. In this review, both the physiology of insulin action and the pathophysiology of insulin resistance are described, focusing on three key insulin target tissues: skeletal muscle, liver, and white adipose tissue. We aim to develop an integrated physiological perspective, placing the intricate signaling effectors that carry out the cell-autonomous response to insulin in the context of the tissue-specific functions that generate the coordinated organismal response. First, in section II, the effectors and effects of direct, cell-autonomous insulin action in muscle, liver, and white adipose tissue are reviewed, beginning at the insulin receptor and working downstream. Section III considers the critical and underappreciated role of tissue crosstalk in whole body insulin action, especially the essential interaction between adipose lipolysis and hepatic gluconeogenesis. The pathophysiology of insulin resistance is then described in section IV. Special attention is given to which signaling pathways and functions become insulin resistant in the setting of chronic overnutrition, and an alternative explanation for the phenomenon of ‟selective hepatic insulin resistanceˮ is presented. Sections V, VI, and VII critically examine the evidence for and against several putative mediators of insulin resistance. Section V reviews work linking the bioactive lipids diacylglycerol, ceramide, and acylcarnitine to insulin resistance; section VI considers the impact of nutrient stresses in the endoplasmic reticulum and mitochondria on insulin resistance; and section VII discusses non-cell autonomous factors proposed to induce insulin resistance, including inflammatory mediators, branched-chain amino acids, adipokines, and hepatokines. Finally, in section VIII, we propose an integrated model of insulin resistance that links these mediators to final common pathways of metabolite-driven gluconeogenesis and ectopic lipid accumulation.
Collapse
Affiliation(s)
- Max C Petersen
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
49
|
Fazakerley DJ, Krycer JR, Kearney AL, Hocking SL, James DE. Muscle and adipose tissue insulin resistance: malady without mechanism? J Lipid Res 2018; 60:1720-1732. [PMID: 30054342 DOI: 10.1194/jlr.r087510] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/25/2018] [Indexed: 12/14/2022] Open
Abstract
Insulin resistance is a major risk factor for numerous diseases, including type 2 diabetes and cardiovascular disease. These disorders have dramatically increased in incidence with modern life, suggesting that excess nutrients and obesity are major causes of "common" insulin resistance. Despite considerable effort, the mechanisms that contribute to common insulin resistance are not resolved. There is universal agreement that extracellular perturbations, such as nutrient excess, hyperinsulinemia, glucocorticoids, or inflammation, trigger intracellular stress in key metabolic target tissues, such as muscle and adipose tissue, and this impairs the ability of insulin to initiate its normal metabolic actions in these cells. Here, we present evidence that the impairment in insulin action is independent of proximal elements of the insulin signaling pathway and is likely specific to the glucoregulatory branch of insulin signaling. We propose that many intracellular stress pathways act in concert to increase mitochondrial reactive oxygen species to trigger insulin resistance. We speculate that this may be a physiological pathway to conserve glucose during specific states, such as fasting, and that, in the presence of chronic nutrient excess, this pathway ultimately leads to disease. This review highlights key points in this pathway that require further research effort.
Collapse
Affiliation(s)
- Daniel J Fazakerley
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia
| | - James R Krycer
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia
| | - Alison L Kearney
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia
| | - Samantha L Hocking
- Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - David E James
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia .,Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
50
|
Xia W, Pessentheiner AR, Hofer DC, Amor M, Schreiber R, Schoiswohl G, Eichmann TO, Walenta E, Itariu B, Prager G, Hackl H, Stulnig T, Kratky D, Rülicke T, Bogner-Strauss JG. Loss of ABHD15 Impairs the Anti-lipolytic Action of Insulin by Altering PDE3B Stability and Contributes to Insulin Resistance. Cell Rep 2018; 23:1948-1961. [PMID: 29768196 PMCID: PMC6390945 DOI: 10.1016/j.celrep.2018.04.055] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 02/26/2018] [Accepted: 04/13/2018] [Indexed: 01/14/2023] Open
Abstract
Elevated circulating fatty acids (FAs) contribute to obesity-associated metabolic complications, but the mechanisms by which insulin suppresses lipolysis are poorly understood. We show that α/β-hydrolase domain-containing 15 (ABHD15) is required for the anti-lipolytic action of insulin in white adipose tissue (WAT). Neither insulin nor glucose treatments can suppress FA mobilization in global and conditional Abhd15-knockout (KO) mice. Accordingly, insulin signaling is impaired in Abhd15-KO adipocytes, as indicated by reduced AKT phosphorylation, glucose uptake, and de novo lipogenesis. In vitro data reveal that ABHD15 associates with and stabilizes phosphodiesterase 3B (PDE3B). Accordingly, PDE3B expression is decreased in the WAT of Abhd15-KO mice, mechanistically explaining increased protein kinase A (PKA) activity, hormone-sensitive lipase (HSL) phosphorylation, and undiminished FA release upon insulin signaling. Ultimately, Abhd15-KO mice develop insulin resistance. Notably, ABHD15 expression is decreased in humans with obesity and diabetes compared to humans with obesity and normal glucose tolerance, identifying ABHD15 as a potential therapeutic target to mitigate insulin resistance.
Collapse
Affiliation(s)
- Wenmin Xia
- Institute of Biochemistry, Graz University of Technology, 8010 Graz, Austria
| | - Ariane R Pessentheiner
- Institute of Biochemistry, Graz University of Technology, 8010 Graz, Austria; Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Dina C Hofer
- Institute of Biochemistry, Graz University of Technology, 8010 Graz, Austria
| | - Melina Amor
- Institute of Biochemistry, Graz University of Technology, 8010 Graz, Austria
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | | | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; Center for Explorative Lipidomics, BioTechMed-Graz, 8010 Graz, Austria
| | - Evelyn Walenta
- Institute of Biochemistry, Graz University of Technology, 8010 Graz, Austria
| | - Bianca Itariu
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Gerhard Prager
- Department of Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Hubert Hackl
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Thomas Stulnig
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Juliane G Bogner-Strauss
- Institute of Biochemistry, Graz University of Technology, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria.
| |
Collapse
|