1
|
Al Madhoun A, Haddad D, Kochumon S, Thomas R, Miranda L, George P, Abu-Khalaf N, Al-Mulla F, Ahmad R. TNF-α/NF-κB mediated upregulation of Dectin-1 in hyperglycemic obesity: implications for metabolic inflammation and diabetes. J Transl Med 2025; 23:462. [PMID: 40270030 PMCID: PMC12016449 DOI: 10.1186/s12967-025-06303-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 02/23/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Dectin-1, a key innate immune receptor, plays a critical role in cellular responses and is implicated in chronic inflammation and metabolic syndromes. This study addresses a pivotal gap in elucidating the regulatory mechanism governing Dectin-1 expressionin obesity and diabetes, hypothesizing that hyperglycemia and TNF-α synergistically upregulate Dectin-1 in adipose tissue (AT), thereby exacerbating inflammatory responses and contributing to metabolic dysfunction. METHODS The study included 95 overweight and obese Kuwaiti individuals, categorized into prediabetic (HbA1c < 6.5%) and diabetic (HbA1c ≥ 6.5%) groups. Anthropometric and clinical measurements were recorded. AT biopsies were obtained for RNA extraction and immunohistochemistry. Pre-adipocytes from lean and obese individuals were cultured, differentiated into adipocytes, and treated with TNF-α under normal or high-glucose conditions to assess Dectin-1 expression. Chromatin immunoprecipitation (ChIP) assays analyzed NF-κB binding to the Dectin-1 promoter. Wildtype and TNF-α-/- mice were used to evaluate TNF-α's effect on Dectin-1 expression in AT. RESULTS Our data demonstrate that hyperglycemic obesity significantly induces Dectin-1 expression in AT through the TNF-α/NF-κB signaling pathway. In a cohort of 95 obese individuals, subdivided into prediabetics (HbA1c < 6.5%, n = 49) and diabetics (HbA1c ≥ 6.5%, n = 46), a strong positive correlation was observed between AT Dectin-1 transcripts and plasma HbA1c levels exclusively in diabetic participants, underscoring the specificity of Dectin-1 upregulation in hyperglycemic conditions. Elevated Dectin-1 expression was consistently associated to increased inflammation markers. Immunohistochemical analysis revealed co-localization and concurrent upregulation of Dectin-1 and TNF-α proteins in hyperglycemic AT. Functional assays in TNF-α deficient mice and human adipocytes further validated that TNF-α and hyperglycemia act cooperatively to regulate Dectin-1 expression. Mechanistically, we demonstrated that NF-κB directly binds to the Dectin-1 promoter, mediating its transcriptional activation in response to glucose and TNF-α. CONCLUSION This study significantly advances the understanding of upregulation Dectin-1 in metabolic inflammation, filling a crucial niche in diabetes research and suggesting new therapeutic targets for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Ashraf Al Madhoun
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Al-Soor Street, P.O. Box 1180, Dasman, 15462, Kuwait.
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait.
| | - Dania Haddad
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait
| | - Shihab Kochumon
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Reeby Thomas
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Lavina Miranda
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Al-Soor Street, P.O. Box 1180, Dasman, 15462, Kuwait
| | - Preethi George
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Al-Soor Street, P.O. Box 1180, Dasman, 15462, Kuwait
| | - Nermeen Abu-Khalaf
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Al-Soor Street, P.O. Box 1180, Dasman, 15462, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| |
Collapse
|
2
|
Das A, Mund C, Hagag E, Garcia-Martin R, Karadima E, Witt A, Peitzsch M, Deussen A, Chavakis T, Noll T, Alexaki VI. Adenylate cyclase 10 promotes brown adipose tissue thermogenesis. iScience 2025; 28:111833. [PMID: 39949963 PMCID: PMC11821413 DOI: 10.1016/j.isci.2025.111833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/01/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Brown adipose tissue (BAT) thermogenesis dissipates energy through heat production and thereby it opposes metabolic disease. It is mediated by mitochondrial membrane uncoupling, yet the mechanisms sustaining the mitochondrial membrane potential (ΔΨm) in brown adipocytes are poorly understood. Here we show that isocitrate dehydrogenase (IDH) activity and the expression of the soluble adenylate cyclase 10 (ADCY10), a CO2/bicarbonate sensor residing in mitochondria, are upregulated in BAT of cold-exposed mice. IDH inhibition or ADCY10 deficiency reduces cold resistance of mice. Mechanistically, IDH increases the ΔΨm in brown adipocytes via ADCY10. ADCY10 sustains complex I activity and the ΔΨm via exchange protein activated by cAMP1 (EPAC1). However, neither IDH nor ADCY10 inhibition affect uncoupling protein 1 (UCP1) expression. Hence, we suggest that ADCY10, acting as a CO2/bicarbonate sensor, mediates the effect of IDH on complex I activity through cAMP-EPAC1 signaling, thereby maintaining the ΔΨm and enabling thermogenesis in brown adipocytes.
Collapse
Affiliation(s)
- Anupam Das
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christine Mund
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Eman Hagag
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ruben Garcia-Martin
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Eleftheria Karadima
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anke Witt
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Mirko Peitzsch
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Andreas Deussen
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thomas Noll
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
3
|
Huynh PM, Wang F, An YA. Hypoxia signaling in the adipose tissue. J Mol Cell Biol 2025; 16:mjae039. [PMID: 39363240 PMCID: PMC11892559 DOI: 10.1093/jmcb/mjae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/12/2024] [Accepted: 10/01/2024] [Indexed: 10/05/2024] Open
Abstract
Obesity per se is rapidly emerging all over the planet and further accounts for many other life-threatening conditions, such as diabetes, cardiovascular diseases, and cancers. Decreased oxygen supply or increased relative oxygen consumption in the adipose tissue results in adipose tissue hypoxia, which is a hallmark of obesity. This review aims to provide an up-to-date overview of the hypoxia signaling in the adipose tissue. First, we summarize literature evidence to demonstrate that hypoxia is regularly observed during adipose tissue remodeling in humans and rodent models with obesity. Next, we discuss how hypoxia-inducible factors (HIFs) are regulated and how adipose tissues behave in response to hypoxia. Then, the differential roles of adipose HIF-1α and HIF-2α in adipose tissue biology and obesity pathology are highlighted. Finally, the review emphasizes the importance of modulating adipose hypoxia as a therapeutic avenue to assist adipose tissues in functionally adapting to hypoxic conditions, ultimately promoting adipose health and improving outcomes due to obesity.
Collapse
Affiliation(s)
- Phu M Huynh
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fenfen Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
4
|
Wang L, Li J, Tang P, Zhu D, Tai L, Wang Y, Miyata T, Woodgett JR, Di LJ. GSK3β Deficiency Expands Obese Adipose Vasculature to Mitigate Metabolic Disorders. Circ Res 2025; 136:91-111. [PMID: 39629559 DOI: 10.1161/circresaha.124.325187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Maintaining a well-developed vascular system alongside adipose tissue (AT) expansion significantly reduces the risk of metabolic complications. Although GSK3β (glycogen synthase kinase-3 beta) is known for its role in various cellular processes, its specific functions in AT and regulation of body homeostasis have not been reported. METHODS GSK3β-floxed and GSK3α-floxed mice were crossed with adiponectin-Cre mice to generate GSK3β or GSK3α adipocyte-specific knockout mice (GSK3βADKO and GSK3αADKO). A comprehensive whole-body metabolism analysis was performed on obese GSK3βADKO mice induced by a high-fat diet. RNA sequencing was conducted on AT of both obese GSK3βADKO and GSK3αADKO mice. Various analyses, including vessel perfusion studies, lipolysis analysis, multiplex protein assays, in vitro protein phosphorylation assays, and whole-mount histology staining, were performed on AT of obese GSK3βADKO mice. Tube-formation experiments were performed using 3B-11 endothelial cells cultured in the conditional medium of matured adipocytes under hypoxic conditions. Chromatin precipitation and immunofluorescence studies were conducted using cultured adipocytes with GSK3 inhibition. RESULTS Our findings provide the first evidence that adipocyte-specific knockout of GSK3β expands AT vascularization and mitigates obesity-related metabolic disorders. GSK3β deficiency, but not GSK3α, in adipocytes activates AMPK (AMP-activated protein kinase), leading to increased phosphorylation and nuclear accumulation of HIF-2α, resulting in enhanced transcriptional regulation. Consequently, adipocytes increased VEGF (vascular endothelial growth factor) expression, which engages VEGFR2 on endothelial cells, promoting angiogenesis, expanding the vasculature, and improving vessel perfusion within obese AT. GSK3β deficiency promotes AT remodeling, shifting unhealthy adipocyte function toward a healthier state by increasing insulin-sensitizing hormone adiponectin and preserving healthy adipocyte function. These effects lead to reduced fibrosis, reactive oxygen species, and ER (endoplasmic reticulum) stress in obese AT and improve metabolic disorders associated with obesity. CONCLUSIONS Deletion of GSK3β in adipocytes activates the AMPK/HIF-2α/VEGF/VEGFR2 axis, promoting vasculature expansion within obese AT. This results in a significantly improved local microenvironment, reducing inflammation and effectively ameliorating metabolic disorders associated with obesity.
Collapse
Affiliation(s)
- Li Wang
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
- The Ministry of Education Frontiers Science Center for Precision Oncology (L.W., L.D.), University of Macau, China
- Proteomics, Metabolomics and Drug development core facility, Faculty of Health Sciences (L.W.), University of Macau, China
| | - Jiajia Li
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
| | - Ping Tang
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
| | - Dongliang Zhu
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
| | - Lixin Tai
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
| | - Yuan Wang
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
| | - Tsukiko Miyata
- Lunenfeld-Tanenbaum Research Institute, Sinai Health and Department of Medical Biophysics, University of Toronto, Ontario, Canada (T.M., J.R.W.)
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health and Department of Medical Biophysics, University of Toronto, Ontario, Canada (T.M., J.R.W.)
| | - Li-Jun Di
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
- The Ministry of Education Frontiers Science Center for Precision Oncology (L.W., L.D.), University of Macau, China
| |
Collapse
|
5
|
Kang GS, Kim YE, Oh HR, Jo HJ, Bok S, Jeon YK, Cheon GJ, Roh TY, Chang YT, Park DJ, Ahn GO. Hypoxia-inducible factor-1α-deficient adipose-tissue macrophages produce the heat to mediate lipolysis of white adipose tissue through uncoupling protein-1. Lab Anim Res 2024; 40:37. [PMID: 39473019 PMCID: PMC11523771 DOI: 10.1186/s42826-024-00224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Uncoupling protein 1 (UCP1) is a proton uncoupler located across the mitochondrial membrane generally involved in thermogenesis of brown adipose tissues. Although UCP1 is known to be strongly expressed in brown adipocytes, recent evidence suggest that white adipocytes can also express UCP1 under certain circumstances such as cold- or β-adrenergic receptor-stimulation, allowing them to acquire brown adipocyte-like features thereby becoming 'beige' adipocytes. RESULTS In this study, we report that UCP1 can be expressed in adipose-tissue macrophages (ATM) lacking functional hypoxia-inducible factor-1 (HIF-1) and this does not require cold- nor β-adrenergic receptor activation. By using myeloid-specific Hif-1α knockout (KO) mice, we observed that these mice were protected from diet-induced obesity and exhibited an improved thermogenic tolerance upon cold challenge. ATM isolated from white adipose tissues (WAT) of these mice fed with high fat diet exhibited significantly higher M2-polarization, decreased glycolysis, increased mitochondrial functions and acetyl-CoA levels, along with increased expression of Ucp1, peroxisome proliferator activated receptor-gamma co-activator-1a, and others involved in histone acetylation. Consistent with the increased Ucp1 gene expression, these ATM produced a significant amount of heat mediating lipolysis of co-cultured adipocytes liberating free fatty acid. Treating ATM with acetate, a substrate for acetyl-CoA synthesis was able to boost the heat production in wild-type or Hif-1α-deficient but not UCP1-deficient macrophages, indicating that UCP1 was necessary for the heat production in macrophages. Lastly, we observed a significant inverse correlation between the number of UCP1-expressing ATM in WAT and the body mass index of human individuals. CONCLUSIONS UCP1-expressing ATM produce the heat to mediate lipolysis of adipocytes, indicating that this can be a novel strategy to treat and prevent diet-induced obesity.
Collapse
Affiliation(s)
- Gi-Sue Kang
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Young-Eun Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Ho Rim Oh
- Department of Nuclear Medicine, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - Hye-Ju Jo
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Seoyeon Bok
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Yoon Kyung Jeon
- Department of Pathology, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, College of Medicine, Seoul National University, Seoul, 03080, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea
- College of Medicine, Cancer Research Institute, Seoul National University, Seoul, 03080, Korea
| | - Tae-Young Roh
- Department of Life Sciences, Ewha Womans University, Seoul, 03760, Korea
| | | | - Do Joong Park
- Department of Surgery, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - G-One Ahn
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea.
- College of Medicine, Cancer Research Institute, Seoul National University, Seoul, 03080, Korea.
| |
Collapse
|
6
|
Wang R, Gomez Salazar M, Pruñonosa Cervera I, Coutts A, French K, Pinto MM, Gohlke S, García-Martín R, Blüher M, Schofield CJ, Kourtzelis I, Stimson RH, Bénézech C, Christian M, Schulz TJ, Gudmundsson EF, Jennings LL, Gudnason VG, Chavakis T, Morton NM, Emilsson V, Michailidou Z. Adipocyte deletion of the oxygen-sensor PHD2 sustains elevated energy expenditure at thermoneutrality. Nat Commun 2024; 15:7483. [PMID: 39209825 PMCID: PMC11362468 DOI: 10.1038/s41467-024-51718-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Enhancing thermogenic brown adipose tissue (BAT) function is a promising therapeutic strategy for metabolic disease. However, predominantly thermoneutral modern human living conditions deactivate BAT. We demonstrate that selective adipocyte deficiency of the oxygen-sensor HIF-prolyl hydroxylase (PHD2) gene overcomes BAT dormancy at thermoneutrality. Adipocyte-PHD2-deficient mice maintain higher energy expenditure having greater BAT thermogenic capacity. In human and murine adipocytes, a PHD inhibitor increases Ucp1 levels. In murine brown adipocytes, antagonising the major PHD2 target, hypoxia-inducible factor-(HIF)-2a abolishes Ucp1 that cannot be rescued by PHD inhibition. Mechanistically, PHD2 deficiency leads to HIF2 stabilisation and binding of HIF2 to the Ucp1 promoter, thus enhancing its expression in brown adipocytes. Serum proteomics analysis of 5457 participants in the deeply phenotyped Age, Gene and Environment Study reveal that serum PHD2 associates with increased risk of metabolic disease. Here we show that adipose-PHD2-inhibition is a therapeutic strategy for metabolic disease and identify serum PHD2 as a disease biomarker.
Collapse
Affiliation(s)
- Rongling Wang
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mario Gomez Salazar
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Iris Pruñonosa Cervera
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Amanda Coutts
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Karen French
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Marlene Magalhaes Pinto
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Sabrina Gohlke
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Ruben García-Martín
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC (CNB-CSIC), Campus-UAM, Madrid, Spain
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research University of Oxford, Oxford, UK
| | - Ioannis Kourtzelis
- Hull York Medical School, York Biomedical Research Institute, University of York, York, UK
| | - Roland H Stimson
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mark Christian
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | | | - Lori L Jennings
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Vilmundur G Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Triantafyllos Chavakis
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Nicholas M Morton
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Valur Emilsson
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Zoi Michailidou
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK.
| |
Collapse
|
7
|
Chaurasiya V, Nidhina Haridas PA, Olkkonen VM. Adipocyte-endothelial cell interplay in adipose tissue physiology. Biochem Pharmacol 2024; 222:116081. [PMID: 38408682 DOI: 10.1016/j.bcp.2024.116081] [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: 10/31/2023] [Revised: 02/02/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Adipose tissue (AT) expansion through hyperplasia or hypertrophy requires vascular remodeling that involves angiogenesis. There is quite some evidence that obese white AT (WAT) displays altered vasculature. Some studies suggest that this is associated with hypoxia, which is thought to play a role in inducing inflammatory activation of the excessively expanding WAT. Increasing evidence, based on genetic manipulations or treatments with inhibitory or activator pharmaceuticals, demonstrates that AT angiogenesis is crucial for AT metabolic function, and thereby for whole body metabolism and metabolic health. Despite some contradiction between studies, disturbance of WAT angiogenesis in obesity could be an important factor driving WAT dysfunction and the comorbidities of obesity. Endothelial cells (ECs) contribute to healthy WAT metabolism via transport of fatty acids and other plasma components, secretory signaling molecules, and extracellular vesicles (EVs). This communication is crucial for adipocyte metabolism and underscores the key role that the AT endothelium plays in systemic energy homeostasis and healthy metabolism. Adipocytes communicate towards the neighboring endothelium through several mechanisms. The pro-inflammatory status of hypertrophic adipocytes in obesity is reflected in ECs activation, which promotes chronic inflammation. On the other hand, adiponectin secreted by the adipocytes is important for healthy endothelial function, and adipocytes also secrete other pro- or anti-angiogenic effector molecules and a wealth of EVs - however, their detailed roles in signaling towards the endothelium are yet poorly understood. To conclude, targeting AT angiogenesis and promoting the healthy communication between adipocytes and ECs represent potentially promising strategies to treat obesity and its comorbidities.
Collapse
Affiliation(s)
- Vaishali Chaurasiya
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland
| | - P A Nidhina Haridas
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland.
| |
Collapse
|
8
|
Yao T, Wei D, Tian X, Zhao L, Wan Q, Zhang X, Cai J, Li S, Diao B, Feng S, Shan B, Shao M, Wu Y. PDGFRβ + cell HIF2α is dispensable for white adipose tissue metabolic remodeling and hepatic lipid accumulation in obese mice. Lipids Health Dis 2024; 23:81. [PMID: 38509584 PMCID: PMC10953078 DOI: 10.1186/s12944-024-02069-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Obesity is associated with extensive white adipose tissue (WAT) expansion and remodeling. Healthy WAT expansion contributes to the maintenance of energy balance in the liver, thereby ameliorating obesity-related hepatic steatosis. Tissue-resident mesenchymal stromal cell populations, including PDGFRβ + perivascular cells, are increasingly recognized pivotal as determinants of the manner in which WAT expands. However, the full array of regulatory factors controlling WAT stromal cell functions remains to be fully elucidated. Hypoxia-inducible factors (HIFs) are critical regulators in WAT stromal cell populations such as adipocyte precursor cells (APCs). It is revealed that HIF1α activation within PDGFRβ + stromal cells results in the suppression of de novo adipogenesis and the promotion of a pro-fibrogenic cellular program in obese animals. However, the role of HIF2α in PDGFRβ + cells remains undetermined in vivo. METHODS New genetic models were employed in which HIF1α (encoded by the Hif1a gene) and HIF2α (encoded by the Epas1 gene) are selectively inactivated in PDGFRβ + cells in an inducible manner using tamoxifen (TAM). With these models, both in vitro and in vivo functional analysis of PDGFRβ + cells lacking HIF proteins were performed. Additionally, comprehensive metabolic phenotyping in diet-induced mouse models were performed to investigate the roles of PDGFRβ + cell HIF proteins in WAT remodeling, liver energy balance and systemic metabolism. RESULTS Unlike HIF1α inactivation, the new findings in this study suggest that inducible ablation of HIF2α in PDGFRβ + cells does not cause apparent effects on WAT expansion induced by obesogenic diet. The adipogenic ability of PDGFRβ + APCs is not significantly altered by genetic HIF2α ablation. Moreover, no difference of key parameters associated with healthy WAT remodeling such as improvements of WAT insulin sensitivity, reduction in metabolic inflammation, as well as changes in liver fat accumulation or systemic glucose metabolism, is detected in PDGFRβ + cell Epas1-deficient mice. CONCLUSION The new findings in this study support that, in contrast to HIF1α, PDGFRβ + cell HIF2α appears dispensable for WAT metabolic remodeling and the resulting effects on liver metabolic homeostasis in diet-induced obesity, underscoring the isoform-specific roles of HIFα proteins in the regulation of adipose tissue biology.
Collapse
Affiliation(s)
- Tao Yao
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Danni Wei
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin Tian
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Lin Zhao
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Qiangyou Wan
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoli Zhang
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Juan Cai
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Siqi Li
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bowen Diao
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Suihan Feng
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Bo Shan
- Cancer Center, Zhejiang University, Hangzhou, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Mengle Shao
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China.
| | - Ying Wu
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
9
|
Abbasi K, Zarezadeh R, Valizadeh A, Mehdizadeh A, Hamishehkar H, Nouri M, Darabi M. White-brown adipose tissue interplay in polycystic ovary syndrome: Therapeutic avenues. Biochem Pharmacol 2024; 220:116012. [PMID: 38159686 DOI: 10.1016/j.bcp.2023.116012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
This study highlights the therapeutic potential of activating brown adipose tissue (BAT) for managing polycystic ovary syndrome (PCOS), a prevalent endocrine disorder associated with metabolic and reproductive abnormalities. BAT plays a crucial role in regulating energy expenditure and systemic insulin sensitivity, making it an attractive target for the treatment of obesity and metabolic diseases. Recent research suggests that impaired BAT function and mass may contribute to the link between metabolic disturbances and reproductive issues in PCOS. Additionally, abnormal white adipose tissue (WAT) can exacerbate these conditions by releasing adipokines and nonesterified fatty acids. In this review, we explored the impact of WAT changes on BAT function in PCOS and discussed the potential of BAT activation as a therapeutic strategy to improve PCOS symptoms. We propose that BAT activation holds promise for managing PCOS; however, further research is needed to confirm its efficacy and to develop clinically feasible methods for BAT activation.
Collapse
Affiliation(s)
- Khadijeh Abbasi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Valizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, Germany.
| |
Collapse
|
10
|
Zhao Y, Xiong W, Li C, Zhao R, Lu H, Song S, Zhou Y, Hu Y, Shi B, Ge J. Hypoxia-induced signaling in the cardiovascular system: pathogenesis and therapeutic targets. Signal Transduct Target Ther 2023; 8:431. [PMID: 37981648 PMCID: PMC10658171 DOI: 10.1038/s41392-023-01652-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 11/21/2023] Open
Abstract
Hypoxia, characterized by reduced oxygen concentration, is a significant stressor that affects the survival of aerobic species and plays a prominent role in cardiovascular diseases. From the research history and milestone events related to hypoxia in cardiovascular development and diseases, The "hypoxia-inducible factors (HIFs) switch" can be observed from both temporal and spatial perspectives, encompassing the occurrence and progression of hypoxia (gradual decline in oxygen concentration), the acute and chronic manifestations of hypoxia, and the geographical characteristics of hypoxia (natural selection at high altitudes). Furthermore, hypoxia signaling pathways are associated with natural rhythms, such as diurnal and hibernation processes. In addition to innate factors and natural selection, it has been found that epigenetics, as a postnatal factor, profoundly influences the hypoxic response and progression within the cardiovascular system. Within this intricate process, interactions between different tissues and organs within the cardiovascular system and other systems in the context of hypoxia signaling pathways have been established. Thus, it is the time to summarize and to construct a multi-level regulatory framework of hypoxia signaling and mechanisms in cardiovascular diseases for developing more therapeutic targets and make reasonable advancements in clinical research, including FDA-approved drugs and ongoing clinical trials, to guide future clinical practice in the field of hypoxia signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Weidong Xiong
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China
| | - Chaofu Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - Junbo Ge
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
11
|
Kang GS, Jo HJ, Lee YR, Oh T, Park HJ, Ahn GO. Sensing the oxygen and temperature in the adipose tissues - who's sensing what? Exp Mol Med 2023; 55:2300-2307. [PMID: 37907745 PMCID: PMC10689767 DOI: 10.1038/s12276-023-01113-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 11/02/2023] Open
Abstract
Adipose tissues, composed of various cell types, including adipocytes, endothelial cells, neurons, and immune cells, are organs that are exposed to dynamic environmental challenges. During diet-induced obesity, white adipose tissues experience hypoxia due to adipocyte hypertrophy and dysfunctional vasculature. Under these conditions, cells in white adipose tissues activate hypoxia-inducible factor (HIF), a transcription factor that activates signaling pathways involved in metabolism, angiogenesis, and survival/apoptosis to adapt to such an environment. Exposure to cold or activation of the β-adrenergic receptor (through catecholamines or chemicals) leads to heat generation, mainly in brown adipose tissues through activating uncoupling protein 1 (UCP1), a proton uncoupler in the inner membrane of the mitochondria. White adipose tissues can undergo a similar process under this condition, a phenomenon known as 'browning' of white adipose tissues or 'beige adipocytes'. While UCP1 expression has largely been confined to adipocytes, HIF can be expressed in many types of cells. To dissect the role of HIF in specific types of cells during diet-induced obesity, researchers have generated tissue-specific knockout (KO) mice targeting HIF pathways, and many studies have commonly revealed that intact HIF-1 signaling in adipocytes and adipose tissue macrophages exacerbates tissue inflammation and insulin resistance. In this review, we highlight some of the key findings obtained from these transgenic mice, including Ucp1 KO mice and other models targeting the HIF pathway in adipocytes, macrophages, or endothelial cells, to decipher their roles in diet-induced obesity.
Collapse
Affiliation(s)
- Gi-Sue Kang
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Hye-Ju Jo
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Ye-Rim Lee
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Taerim Oh
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Hye-Joon Park
- College of Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - G-One Ahn
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea.
- College of Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea.
| |
Collapse
|
12
|
Witt A, Mateska I, Palladini A, Sinha A, Wölk M, Harauma A, Bechmann N, Pamporaki C, Dahl A, Rothe M, Kopaliani I, Adolf C, Riester A, Wielockx B, Bornstein SR, Kroiss M, Peitzsch M, Moriguchi T, Fedorova M, Grzybek M, Chavakis T, Mirtschink P, Alexaki VI. Fatty acid desaturase 2 determines the lipidomic landscape and steroidogenic function of the adrenal gland. SCIENCE ADVANCES 2023; 9:eadf6710. [PMID: 37478183 PMCID: PMC10361602 DOI: 10.1126/sciadv.adf6710] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 06/16/2023] [Indexed: 07/23/2023]
Abstract
Corticosteroids regulate vital processes, including stress responses, systemic metabolism, and blood pressure. Here, we show that corticosteroid synthesis is related to the polyunsaturated fatty acid (PUFA) content of mitochondrial phospholipids in adrenocortical cells. Inhibition of the rate-limiting enzyme of PUFA synthesis, fatty acid desaturase 2 (FADS2), leads to perturbations in the mitochondrial lipidome and diminishes steroidogenesis. Consistently, the adrenocortical mitochondria of Fads2-/- mice fed a diet with low PUFA concentration are structurally impaired and corticoid levels are decreased. On the contrary, FADS2 expression is elevated in the adrenal cortex of obese mice, and plasma corticosterone is increased, which can be counteracted by dietary supplementation with the FADS2 inhibitor SC-26192 or icosapent ethyl, an eicosapentaenoic acid ethyl ester. In humans, FADS2 expression is elevated in aldosterone-producing adenomas compared to non-active adenomas or nontumorous adrenocortical tissue and correlates with expression of steroidogenic genes. Our data demonstrate that FADS2-mediated PUFA synthesis determines adrenocortical steroidogenesis in health and disease.
Collapse
Affiliation(s)
- Anke Witt
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Ivona Mateska
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Alessandra Palladini
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Centre Munich at the University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, 85764, Germany
| | - Anupam Sinha
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Michele Wölk
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Akiko Harauma
- School of Life and Environmental Science, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa, 252-5201, Japan
| | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Christina Pamporaki
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Andreas Dahl
- DRESDEN-Concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, 01307, Germany
| | | | - Irakli Kopaliani
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Christian Adolf
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, Munich, 80336, Germany
| | - Anna Riester
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, Munich, 80336, Germany
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Stefan R. Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Matthias Kroiss
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, Munich, 80336, Germany
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, Wuerzburg, 97080, Germany
| | - Mirko Peitzsch
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Toru Moriguchi
- School of Life and Environmental Science, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa, 252-5201, Japan
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Michal Grzybek
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Centre Munich at the University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, 85764, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Peter Mirtschink
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| |
Collapse
|
13
|
Trevaskis NL, Martinez-Corral I, García-Caballero M. Editorial: Modulating lymphatic vascular growth in disease: current and potential pharmacological approaches for prevention and treatment, Volume II. Front Pharmacol 2023; 14:1224414. [PMID: 37469868 PMCID: PMC10352829 DOI: 10.3389/fphar.2023.1224414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/23/2023] [Indexed: 07/21/2023] Open
Affiliation(s)
- Natalie L. Trevaskis
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Ines Martinez-Corral
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, Inserm UMR-S1172, University of Lille, Inserm, CHU Lille, Lille, France
| | - Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Andalucía Tech, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| |
Collapse
|
14
|
Sun K, Li X, Scherer PE. Extracellular Matrix (ECM) and Fibrosis in Adipose Tissue: Overview and Perspectives. Compr Physiol 2023; 13:4387-4407. [PMID: 36715281 PMCID: PMC9957663 DOI: 10.1002/cphy.c220020] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fibrosis in adipose tissue is a major driver of obesity-related metabolic dysregulation. It is characterized by an overaccumulation of extracellular matrix (ECM) during unhealthy expansion of adipose tissue in response to over nutrition. In obese adipose-depots, hypoxia stimulates multiple pro-fibrotic signaling pathways in different cell populations, thereby inducing the overproduction of the ECM components, including collagens, noncollagenous proteins, and additional enzymatic components of ECM synthesis. As a consequence, local fibrosis develops. The result of fibrosis-induced mechanical stress not only triggers cell necrosis and inflammation locally in adipose tissue but also leads to system-wide lipotoxicity and insulin resistance. A better understanding of the mechanisms underlying the obesity-induced fibrosis will help design therapeutic approaches to reduce or reverse the pathological changes associated with obese adipose tissue. Here, we aim to summarize the major advances in the field, which include newly identified fibrotic factors, cell populations that contribute to the fibrosis in adipose tissue, as well as novel mechanisms underlying the development of fibrosis. We further discuss the potential therapeutic strategies to target fibrosis in adipose tissue for the treatment of obesity-linked metabolic diseases and cancer. © 2023 American Physiological Society. Compr Physiol 13:4387-4407, 2023.
Collapse
Affiliation(s)
- Kai Sun
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xin Li
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Philipp E. Scherer
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
15
|
Al Madhoun A, Kochumon S, Al-Rashed F, Sindhu S, Thomas R, Miranda L, Al-Mulla F, Ahmad R. Dectin-1 as a Potential Inflammatory Biomarker for Metabolic Inflammation in Adipose Tissue of Individuals with Obesity. Cells 2022; 11:2879. [PMID: 36139454 PMCID: PMC9496833 DOI: 10.3390/cells11182879] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/02/2022] [Accepted: 09/10/2022] [Indexed: 11/17/2022] Open
Abstract
In obesity, macrophage activation and infiltration in adipose tissue (AT) underlie chronic low-grade inflammation-induced insulin resistance. Although dectin-1 is primarily a pathogen recognition receptor and innate immune response modulator, its role in metabolic syndromes remains to be clarified. This study aimed to investigate the dectin-1 gene expression in subcutaneous AT in the context of obesity and associated inflammatory markers. Subcutaneous AT biopsies were collected from 59 nondiabetic (lean/overweight/obese) individuals. AT gene expression levels of dectin-1 and inflammatory markers were determined via real-time reverse transcriptase-quantitative polymerase chain reaction. Dectin-1 protein expression was assessed using immunohistochemistry. Plasma lipid profiles were measured by ELISA. AT dectin-1 transcripts and proteins were significantly elevated in obese as compared to lean individuals. AT dectin-1 transcripts correlated positively with body mass index and fat percentage (r ≥ 0.340, p ≤ 0.017). AT dectin-1 RNA levels correlated positively with clinical parameters, including plasma C-reactive protein and CCL5/RANTES, but negatively with that of adiponectin. The expression of dectin-1 transcripts was associated with that of various proinflammatory cytokines, chemokines, and their cognate receptors (r ≥ 0.300, p ≤ 0.05), but not with anti-inflammatory markers. Dectin-1 and members of the TLR signaling cascade were found to be significantly associated, suggesting an interplay between the two pathways. Dectin-1 expression was correlated with monocyte/macrophage markers, including CD16, CD68, CD86, and CD163, suggesting its monocytes/macrophage association in an adipose inflammatory microenvironment. Dectin-1 expression was independently predicted by CCR5, CCL20, TLR2, and MyD88. In conclusion, dectin-1 may be regarded as an AT biomarker of metabolic inflammation in obesity.
Collapse
Affiliation(s)
- Ashraf Al Madhoun
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Shihab Kochumon
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fatema Al-Rashed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Sardar Sindhu
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Reeby Thomas
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Lavina Miranda
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| |
Collapse
|
16
|
Moon JS, Riopel M, Seo JB, Herrero-Aguayo V, Isaac R, Lee YS. HIF-2α Preserves Mitochondrial Activity and Glucose Sensing in Compensating β-Cells in Obesity. Diabetes 2022; 71:1508-1524. [PMID: 35472707 PMCID: PMC9233300 DOI: 10.2337/db21-0736] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 04/08/2022] [Indexed: 11/13/2022]
Abstract
In obesity, increased mitochondrial metabolism with the accumulation of oxidative stress leads to mitochondrial damage and β-cell dysfunction. In particular, β-cells express antioxidant enzymes at relatively low levels and are highly vulnerable to oxidative stress. Early in the development of obesity, β-cells exhibit increased glucose-stimulated insulin secretion in order to compensate for insulin resistance. This increase in β-cell function under the condition of enhanced metabolic stress suggests that β-cells possess a defense mechanism against increased oxidative damage, which may become insufficient or decline at the onset of type 2 diabetes. Here, we show that metabolic stress induces β-cell hypoxia inducible factor 2α (HIF-2α), which stimulates antioxidant gene expression (e.g., Sod2 and Cat) and protects against mitochondrial reactive oxygen species (ROS) and subsequent mitochondrial damage. Knockdown of HIF-2α in Min6 cells exaggerated chronic high glucose-induced mitochondrial damage and β-cell dysfunction by increasing mitochondrial ROS levels. Moreover, inducible β-cell HIF-2α knockout mice developed more severe β-cell dysfunction and glucose intolerance on a high-fat diet, along with increased ROS levels and decreased islet mitochondrial mass. Our results provide a previously unknown mechanism through which β-cells defend against increased metabolic stress to promote β-cell compensation in obesity.
Collapse
Affiliation(s)
- Jae-Su Moon
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Matthew Riopel
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Jong Bae Seo
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Vicente Herrero-Aguayo
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
| | - Roi Isaac
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Yun Sok Lee
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
- Corresponding author: Yun Sok Lee,
| |
Collapse
|
17
|
Adipocyte HIF2α functions as a thermostat via PKA Cα regulation in beige adipocytes. Nat Commun 2022; 13:3268. [PMID: 35672324 PMCID: PMC9174489 DOI: 10.1038/s41467-022-30925-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 05/24/2022] [Indexed: 12/14/2022] Open
Abstract
Thermogenic adipocytes generate heat to maintain body temperature against hypothermia in response to cold. Although tight regulation of thermogenesis is required to prevent energy sources depletion, the molecular details that tune thermogenesis are not thoroughly understood. Here, we demonstrate that adipocyte hypoxia-inducible factor α (HIFα) plays a key role in calibrating thermogenic function upon cold and re-warming. In beige adipocytes, HIFα attenuates protein kinase A (PKA) activity, leading to suppression of thermogenic activity. Mechanistically, HIF2α suppresses PKA activity by inducing miR-3085-3p expression to downregulate PKA catalytic subunit α (PKA Cα). Ablation of adipocyte HIF2α stimulates retention of beige adipocytes, accompanied by increased PKA Cα during re-warming after cold stimuli. Moreover, administration of miR-3085-3p promotes beige-to-white transition via downregulation of PKA Cα and mitochondrial abundance in adipocyte HIF2α deficient mice. Collectively, these findings suggest that HIF2α-dependent PKA regulation plays an important role as a thermostat through dynamic remodeling of beige adipocytes. Thermogenic adipocytes maintain body temperature in response to cold, but how this is tuned during cold and re-warming is unclear. Here, the authors show HIF2α inhibits beige adipocyte retention, regulating PKA catalysis to control dynamic adipocyte remodelling.
Collapse
|
18
|
Song L, Cao X, Ji W, Zhao L, Yang W, Lu M, Yang J. Inhibition of STAT3 enhances UCP1 expression and mitochondrial function in brown adipocytes. Eur J Pharmacol 2022; 926:175040. [DOI: 10.1016/j.ejphar.2022.175040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/04/2022] [Accepted: 05/16/2022] [Indexed: 11/03/2022]
|
19
|
Jiang X, Tian W, Kim D, McQuiston AS, Vinh R, Rockson SG, Semenza GL, Nicolls MR. Hypoxia and Hypoxia-Inducible Factors in Lymphedema. Front Pharmacol 2022; 13:851057. [PMID: 35450048 PMCID: PMC9017680 DOI: 10.3389/fphar.2022.851057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/14/2022] [Indexed: 12/19/2022] Open
Abstract
Lymphedema is a chronic inflammatory disorder characterized by edema, fat deposition, and fibrotic tissue remodeling. Despite significant advances in lymphatic biology research, our knowledge of lymphedema pathology is incomplete. Currently, there is no approved pharmacological therapy for this debilitating disease. Hypoxia is a recognized feature of inflammation, obesity, and fibrosis. Understanding hypoxia-regulated pathways in lymphedema may provide new insights into the pathobiology of this chronic disorder and help develop new medicinal treatments.
Collapse
Affiliation(s)
- Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| | - Dongeon Kim
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| | - Alexander S McQuiston
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| | - Ryan Vinh
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| | | | - Gregg L Semenza
- Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry, and McKusick-Nathans Institute of Genetic Medicine, Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark R Nicolls
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
20
|
Barrea L, Caprio M, Watanabe M, Cammarata G, Feraco A, Muscogiuri G, Verde L, Colao A, Savastano S. Could very low-calorie ketogenic diets turn off low grade inflammation in obesity? Emerging evidence. Crit Rev Food Sci Nutr 2022; 63:8320-8336. [PMID: 35373658 DOI: 10.1080/10408398.2022.2054935] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Obesity is an emerging non-communicable disease associated with chronic low-grade inflammation and oxidative stress, compounded by the development of many obesity-related diseases, such as cardiovascular disease, type 2 diabetes mellitus, and a range of cancers. Originally developed for the treatment of epilepsy in drug non-responder children, the ketogenic diet (KD) is being increasingly used in the treatment of many diseases, including obesity and obesity-related conditions. The KD is a dietary pattern characterized by high fat intake, moderate to low protein consumption, and very low carbohydrate intake (<50 g) that has proved to be an effective and weight-loss tool. In addition, it also appears to be a dietary intervention capable of improving the inflammatory state and oxidative stress in individuals with obesity by means of several mechanisms. The main activity of the KD has been linked to improving mitochondrial function and decreasing oxidative stress. β-hydroxybutyrate, the most studied ketone body, has been shown to reduce the production of reactive oxygen species, improving mitochondrial respiration. In addition, KDs exert anti-inflammatory activity through several mechanisms, e.g., by inhibiting activation of the nuclear factor kappa-light-chain-enhancer of activated B cells, and the inflammatory nucleotide-binding, leucine-rich-containing family, pyrin domain-containing-3, and inhibiting histone deacetylases. Given the rising interest in the topic, this review looks at the underlying anti-inflammatory and antioxidant mechanisms of KDs and their possible recruitment in the treatment of obesity and obesity-related disorders.
Collapse
Affiliation(s)
- Luigi Barrea
- Dipartimento di Scienze Umanistiche, Università Telematica Pegaso, Napoli, Italy
- Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| | - Mikiko Watanabe
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Cammarata
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI) and Center of Excellence for Biomedical Research, University of Genova, Genova, Italy
| | - Alessandra Feraco
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| | - Giovanna Muscogiuri
- Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Naples, Italy
- Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile", University Federico II, Naples, Italy
| | - Ludovica Verde
- Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Naples, Italy
| | - Annamaria Colao
- Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Naples, Italy
- Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile", University Federico II, Naples, Italy
| | - Silvia Savastano
- Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Naples, Italy
| |
Collapse
|
21
|
Zheng X, Ma J, Hu M, Long J, Wei Q, Ren W. Analysis of HIF2α polymorphisms in infertile women with polycystic ovary syndrome or unexplained infertility. Front Endocrinol (Lausanne) 2022; 13:986567. [PMID: 36157441 PMCID: PMC9492870 DOI: 10.3389/fendo.2022.986567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/17/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE To evaluate HIF2α polymorphisms and glucose metabolism in a group of women with polycystic ovary syndrome (PCOS) or unexplained infertility (UI). PATIENTS The infertile group (n=148) consisted of 96 women with PCOS, 52 women with UI, and176 women without infertility as a healthy control group. INTERVENTION We genotyped 29 single nucleotide polymorphisms (SNPs) of HIF2α by using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS)-based genotyping technology. The genetic associations were analyzed statistically. MAIN OUTCOME MEASURES Allele frequency, genotype distribution and haplotype analyze of the HIF2α polymorphisms were performed. Body mass index (BMI), waist circumference, uric acid (UA), high-sensitivity C-reactive protein (hsCRP), lipids, glucose and insulin tolerance - were also measured. RESULTS Infertile women with PCOS had a higherBMI and waist circumference, elevated hsCRP and uric acid (UA) levels, impaired glucose tolerance, and increased levels of plasma insulin compared to UI patients and healthy women. SNP analysis of HIF2α revealed that the allele and genotype frequencies of rs4953361 were significantly associated with infertile women with PCOS. Haplotype analysis of the HIF2α polymorphism identified haplotypes TGG and TGA as being associated with infertile women with PCOS. Women with the AA genotype of rs4953361 had a significantly higher BMI and post load plasma glucose and insulin levels than those of women with the GG genotype. CONCLUSION Infertile women with PCOS more commonly have metabolic disturbances than those with UI. This is the first study to report an association between HIF2α polymorphisms (rs4953361) and the risk of infertile women with PCOS, not UI, in Han Chinese population. These results require replication in larger populations.In this observational study, we did not report the results of a health care intervention on human participants. The study was approved by the Human Research Ethics Committee of the First Affiliated Hospital of Chongqing Medical University. Clinical data and peripheral blood samples were collected only after explaining the objectives of the study and obtaining a signed informed consent form.
Collapse
Affiliation(s)
- Xiaoya Zheng
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Xiaoya Zheng,
| | - Jiani Ma
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Hu
- Reproductive and Infertility Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Long
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiang Wei
- Prevention of Disease Department, Chongqing Jiulongpo District Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Wei Ren
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Kirasirova EA, Frolkina EA, Rybalchenko IE, Tyutina SI. [Chronic respiratory failure correction in cicatricial laryngeal and tracheal stenosis using helium-oxygen mixture]. Vestn Otorinolaringol 2022; 87:63-70. [PMID: 36107183 DOI: 10.17116/otorino20228704163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The article provides a review of the literature on the development of chronic respiratory failure in patients with chronic cicatricial stenosis of the larynx and cervical trachea. The authors provide data on the etiology, pathogenetic features of the course of cicatricial stenosis of the larynx and trachea, the reasons for the development of chronic respiratory failure, the effect of hypoxemia on general metabolic processes in the body and on regeneration processes, as well as on methods of their correction and improvement of the postoperative period. The methods of respiratory impact on chronic respiratory failure in these patients are considered, based on the experience of a number of researchers and technical advances in recent years.
Collapse
Affiliation(s)
- E A Kirasirova
- Pirogov Russian National Research Medical University, Moscow, Russia
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| | - E A Frolkina
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| | | | - S I Tyutina
- Sverzhevsky Research Clinical Institute of Otorhinolaryngology, Moscow, Russia
| |
Collapse
|
23
|
Bi X, Li Y, Dong Z, Zhao J, Wu W, Zou J, Guo L, Lu F, Gao J. Recent Developments in Extracellular Matrix Remodeling for Fat Grafting. Front Cell Dev Biol 2021; 9:767362. [PMID: 34977018 PMCID: PMC8716396 DOI: 10.3389/fcell.2021.767362] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022] Open
Abstract
Remodeling of the extracellular matrix (ECM), which provides structural and biochemical support for surrounding cells, is vital for adipose tissue regeneration after autologous fat grafting. Rapid and high-quality ECM remodeling can improve the retention rate after fat grafting by promoting neovascularization, regulating stem cells differentiation, and suppressing chronic inflammation. The degradation and deposition of ECM are regulated by various factors, including hypoxia, blood supply, inflammation, and stem cells. By contrast, ECM remodeling alters these regulatory factors, resulting in a dynamic relationship between them. Although researchers have attempted to identify the cellular sources of factors associated with tissue regeneration and regulation of the microenvironment, the factors and mechanisms that affect adipose tissue ECM remodeling remain incompletely understood. This review describes the process of adipose ECM remodeling after grafting and summarizes the factors that affect ECM reconstruction. Also, this review provides an overview of the clinical methods to avoid poor ECM remodeling. These findings may provide new ideas for improving the retention of adipose tissue after fat transplantation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
24
|
B Tóth B, Barta Z, Barta ÁB, Fésüs L. Regulatory modules of human thermogenic adipocytes: functional genomics of large cohort and Meta-analysis derived marker-genes. BMC Genomics 2021; 22:886. [PMID: 34895148 PMCID: PMC8665548 DOI: 10.1186/s12864-021-08126-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 10/27/2021] [Indexed: 11/18/2022] Open
Abstract
Background Recently, ProFAT and BATLAS studies identified brown and white adipocytes marker genes based on analysis of large databases. They offered scores to determine the thermogenic status of adipocytes using the gene-expression data of these markers. In this work, we investigated the functional context of these genes. Results Gene Set Enrichment Analyses (KEGG, Reactome) of the BATLAS and ProFAT marker-genes identified pathways deterministic in the formation of brown and white adipocytes. The collection of the annotated proteins of the defined pathways resulted in expanded white and brown characteristic protein-sets, which theoretically contain all functional proteins that could be involved in the formation of adipocytes. Based on our previously obtained RNA-seq data, we visualized the expression profile of these proteins coding genes and found patterns consistent with the two adipocyte phenotypes. The trajectory of the regulatory processes could be outlined by the transcriptional profile of progenitor and differentiated adipocytes, highlighting the importance of suppression processes in browning. Protein interaction network-based functional genomics by STRING, Cytoscape and R-Igraph platforms revealed that different biological processes shape the brown and white adipocytes and highlighted key regulatory elements and modules including GAPDH-CS, DECR1, SOD2, IL6, HRAS, MTOR, INS-AKT, ERBB2 and 4-NFKB, and SLIT-ROBO-MAPK. To assess the potential role of a particular protein in shaping adipocytes, we assigned interaction network location-based scores (betweenness centrality, number of bridges) to them and created a freely accessible platform, the AdipoNET (https//adiponet.com), to conveniently use these data. The Eukaryote Promoter Database predicted the response elements in the UCP1 promoter for the identified, potentially important transcription factors (HIF1A, MYC, REL, PPARG, TP53, AR, RUNX, and FoxO1). Conclusion Our integrative approach-based results allowed us to investigate potential regulatory elements of thermogenesis in adipose tissue. The analyses revealed that some unique biological processes form the brown and white adipocyte phenotypes, which presumes the existence of the transitional states. The data also suggests that the two phenotypes are not mutually exclusive, and differentiation of thermogenic adipocyte requires induction of browning as well as repressions of whitening. The recognition of these simultaneous actions and the identified regulatory modules can open new direction in obesity research. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08126-8.
Collapse
Affiliation(s)
- Beáta B Tóth
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem Tér 1, Debrecen, H-4032, Hungary.
| | - Zoltán Barta
- MTA-DE Behavioural Ecology Research Group, Department of Evolutionary Zoology and Human Biology, University of Debrecen, Egyetem tér 1, Debrecen, H-4032, Hungary
| | - Ákos Barnabás Barta
- Vienna University of Economics and Business (WU), Welthandelspl. 1, 1020, Wien, Austria
| | - László Fésüs
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem Tér 1, Debrecen, H-4032, Hungary.
| |
Collapse
|
25
|
Ito T, Yamamoto Y, Yamagishi N, Kanai Y. Stomach secretes estrogen in response to the blood triglyceride levels. Commun Biol 2021; 4:1364. [PMID: 34876651 PMCID: PMC8651635 DOI: 10.1038/s42003-021-02901-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 11/23/2021] [Indexed: 11/29/2022] Open
Abstract
Mammals receive body energy information to maintain energy homeostasis. Ghrelin, insulin, leptin and vagal afferents transmit the status of fasting, blood glucose, body fat, and food intake, respectively. Estrogen also inhibits feeding behavior and lipogenesis, but increases body fat mass. However, how blood triglyceride levels are monitored and the physiological roles of estrogen from the perspective of lipid homeostasis remain unsettled. Here, we show that stomach secretes estrogen in response to the blood triglyceride levels. Estrogen-secreting gastric parietal cells predominantly use fatty acids as an energy source. Blood estrogen levels increase as blood triglyceride levels rise in a stomach-dependent manner. Estrogen levels in stomach tissues increase as blood triglyceride levels rise, and isolated gastric gland epithelium produces estrogen in a fatty acid-dependent manner. We therefore propose that stomach monitors and controls blood triglyceride levels using estrogen, which inhibits feeding behavior and lipogenesis, and promotes triglyceride uptake by adipocytes.
Collapse
Affiliation(s)
- Takao Ito
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuta Yamamoto
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naoko Yamagishi
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yoshimitsu Kanai
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
26
|
Essential role of systemic iron mobilization and redistribution for adaptive thermogenesis through HIF2-α/hepcidin axis. Proc Natl Acad Sci U S A 2021; 118:2109186118. [PMID: 34593646 DOI: 10.1073/pnas.2109186118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2021] [Indexed: 12/23/2022] Open
Abstract
Iron is an essential biometal, but is toxic if it exists in excess. Therefore, iron content is tightly regulated at cellular and systemic levels to meet metabolic demands but to avoid toxicity. We have recently reported that adaptive thermogenesis, a critical metabolic pathway to maintain whole-body energy homeostasis, is an iron-demanding process for rapid biogenesis of mitochondria. However, little information is available on iron mobilization from storage sites to thermogenic fat. This study aimed to determine the iron-regulatory network that underlies beige adipogenesis. We hypothesized that thermogenic stimulus initiates the signaling interplay between adipocyte iron demands and systemic iron liberation, resulting in iron redistribution into beige fat. To test this hypothesis, we induced reversible activation of beige adipogenesis in C57BL/6 mice by administering a β3-adrenoreceptor agonist CL 316,243 (CL). Our results revealed that CL stimulation induced the iron-regulatory protein-mediated iron import into adipocytes, suppressed hepcidin transcription, and mobilized iron from the spleen. Mechanistically, CL stimulation induced an acute activation of hypoxia-inducible factor 2-α (HIF2-α), erythropoietin production, and splenic erythroid maturation, leading to hepcidin suppression. Disruption of systemic iron homeostasis by pharmacological HIF2-α inhibitor PT2385 or exogenous administration of hepcidin-25 significantly impaired beige fat development. Our findings suggest that securing iron availability via coordinated interplay between renal hypoxia and hepcidin down-regulation is a fundamental mechanism to activate adaptive thermogenesis. It also provides an insight into the effects of adaptive thermogenesis on systemic iron mobilization and redistribution.
Collapse
|
27
|
Li X, Zhang X, Xia J, Zhang L, Chen B, Lian G, Yun C, Yang J, Yan Y, Wang P, Wang X, Liu B, Liu H, Liang H, Pang Y, Wang X, Jiang C. Macrophage HIF-2α suppresses NLRP3 inflammasome activation and alleviates insulin resistance. Cell Rep 2021; 36:109607. [PMID: 34433035 DOI: 10.1016/j.celrep.2021.109607] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/22/2021] [Accepted: 08/05/2021] [Indexed: 12/13/2022] Open
Abstract
The interrelation between hypoxia and immune response has pivotal roles in the pathogenesis of chronic metabolic diseases. However, the role of macrophage HIF-2α in NLRP3 inflammasome activation remains unclear. Here, we show that deficiency of HIF-2α in macrophages results in excessive activation of the NLRP3 inflammasome in a manner dependent on CPT1A-mediated enhancement of fatty acid oxidation (FAO). Mechanistically, HIF-2α binds directly to the Cpt1a promoter and is involved in the regulation of H3K27me3 methylation during NLRP3 inflammasome activation. Myeloid-specific Hif2α knockout mice exhibit exacerbated insulin resistance and increased activation of NLRP3 inflammasome in macrophages. Overexpression of the Hif2α gene or stabilization of the protein by FG-4592 ameliorates insulin resistance and reduces NLRP3 inflammasome activation in macrophages. Taken together, our results suggest that macrophage HIF-2α inhibits FAO-mediated activation of the NLRP3 inflammasome and alleviates insulin resistance.
Collapse
Affiliation(s)
- Xiaopeng Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Xiujuan Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialin Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Linqi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Bo Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Guan Lian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Chuyu Yun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Juan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yu Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Pengcheng Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Xuemei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Bo Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Huiying Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Hui Liang
- Institute of Systems Biomedicine, School of Basic Medicine, Peking University, Beijing 100191, China
| | - Yanli Pang
- Department of Obstetrics and Gynecology, Third Hospital, Peking University, Beijing, China
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China.
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.
| |
Collapse
|
28
|
Vanderhaeghen T, Beyaert R, Libert C. Bidirectional Crosstalk Between Hypoxia Inducible Factors and Glucocorticoid Signalling in Health and Disease. Front Immunol 2021; 12:684085. [PMID: 34149725 PMCID: PMC8211996 DOI: 10.3389/fimmu.2021.684085] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Glucocorticoid-induced (GC) and hypoxia-induced transcriptional responses play an important role in tissue homeostasis and in the regulation of cellular responses to stress and inflammation. Evidence exists that there is an important crosstalk between both GC and hypoxia effects. Hypoxia is a pathophysiological condition to which cells respond quickly in order to prevent metabolic shutdown and death. The hypoxia inducible factors (HIFs) are the master regulators of oxygen homeostasis and are responsible for the ability of cells to cope with low oxygen levels. Maladaptive responses of HIFs contribute to a variety of pathological conditions including acute mountain sickness (AMS), inflammation and neonatal hypoxia-induced brain injury. Synthetic GCs which are analogous to the naturally occurring steroid hormones (cortisol in humans, corticosterone in rodents), have been used for decades as anti-inflammatory drugs for treating pathological conditions which are linked to hypoxia (i.e. asthma, ischemic injury). In this review, we investigate the crosstalk between the glucocorticoid receptor (GR), and HIFs. We discuss possible mechanisms by which GR and HIF influence one another, in vitro and in vivo, and the therapeutic effects of GCs on HIF-mediated diseases.
Collapse
Affiliation(s)
- Tineke Vanderhaeghen
- Centre for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Rudi Beyaert
- Centre for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Claude Libert
- Centre for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
29
|
Fu P, Zhu R, Jia J, Hu Y, Wu C, Cieszczyk P, Holmberg HC, Gong L. Aerobic exercise promotes the functions of brown adipose tissue in obese mice via a mechanism involving COX2 in the VEGF signaling pathway. Nutr Metab (Lond) 2021; 18:56. [PMID: 34082784 PMCID: PMC8176720 DOI: 10.1186/s12986-021-00581-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 05/27/2021] [Indexed: 12/18/2022] Open
Abstract
Background High-fat diet (HFD)-induced obesity causes immune cells to infiltrate adipose tissue, leading to chronic inflammation and metabolic syndrome. Brown adipose tissue (BAT) can dissipate the energy produced by lipid oxidation as heat, thereby counteracting obesity. Aerobic exercise activates BAT, but the specific underlying mechanism is still unclear. Methods Male C57BL/6 J mice were divided into a normal diet control group (NC group) and HFD group (H group). After becoming obese, the animals in the H group were subdivided into a control group (HC group) and an exercise group (HE group, with treadmill training). After 4 weeks, the mRNA profile of BAT was determined, and then differentially expressed key genes and pathways were verified in vitro. Results Relative to the NC group, the genes upregulated in the HC group coded mainly for proteins involved in immune system progression and inflammatory and immune responses, while the downregulated genes regulated lipid metabolism and oxidation–reduction. Relative to the HC group, the genes upregulated in the HE group coded for glycolipid metabolism, while those that were downregulated were involved in cell death and apoptosis. VEGF and other signaling pathways were enhanced by aerobic exercise. Interaction analysis revealed that the gene encoding cyclooxygenase 2 (COX2) of the VEGF signaling pathway is central to this process, which was verified by a sympathetic activator (isoprenaline hydrochloride) and COX2 inhibitor (NS-398). Conclusions In mice with HFD-induced obesity, four weeks of aerobic exercise elevated BAT mass and increased the expression of genes related to glycolipid metabolism and anti-inflammatory processes. Several pathways are involved, with COX2 in the VEGF signaling pathway playing a key role.
Collapse
Affiliation(s)
- Pengyu Fu
- China Institute of Sport and Health Science, Beijing Sport University, Xinxi Road 48, Haidian District, Beijing, 100084, China.,Department of Physical Education, Northwestern Polytechnical University, West Youyi Road 127, Beilin District, Shaanxi, 710109, China
| | - Rongxin Zhu
- China Institute of Sport and Health Science, Beijing Sport University, Xinxi Road 48, Haidian District, Beijing, 100084, China.,Shanghai Research Institute of Sports Science, Xuhui District, Wuxing Road 87, Shanghai, 200030, China
| | - Jie Jia
- China Institute of Sport and Health Science, Beijing Sport University, Xinxi Road 48, Haidian District, Beijing, 100084, China.,Sport Science College, Beijing Sport University, Xinxi Road 48, Haidian District, Beijing, 100084, China
| | - Yang Hu
- China Institute of Sport and Health Science, Beijing Sport University, Xinxi Road 48, Haidian District, Beijing, 100084, China
| | - Chengjun Wu
- School of Biomedical Engineering, Dalian University of Technology and IC Technology Key Lab of Liaoning, Dalian, 116024, China
| | - Pawel Cieszczyk
- Department of Molecular Biology, Faculty of Physical Education, Gdańsk University of Physical Education and Sport, ul. Kazimierza Górskiego 1, 80-336, Gdańsk, Poland
| | - Hans-Christer Holmberg
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institute, Stockholm, Sweden
| | - Lijing Gong
- China Institute of Sport and Health Science, Beijing Sport University, Xinxi Road 48, Haidian District, Beijing, 100084, China.
| |
Collapse
|
30
|
Moustakas II, Katsarou A, Legaki AI, Pyrina I, Ntostoglou K, Papatheodoridi AM, Gercken B, Pateras IS, Gorgoulis VG, Koutsilieris M, Chavakis T, Chatzigeorgiou A. Hepatic Senescence Accompanies the Development of NAFLD in Non-Aged Mice Independently of Obesity. Int J Mol Sci 2021; 22:3446. [PMID: 33810566 PMCID: PMC8037476 DOI: 10.3390/ijms22073446] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 01/21/2023] Open
Abstract
Senescence is considered to be a cardinal player in several chronic inflammatory and metabolic pathologies. The two dominant mechanisms of senescence include replicative senescence, predominantly depending on age-induced telomere shortening, and stress-induced senescence, triggered by external or intracellular harmful stimuli. Recent data indicate that hepatocyte senescence is involved in the development of nonalcoholic fatty liver disease (NAFLD). However, previous studies have mainly focused on age-related senescence during NAFLD, in the presence or absence of obesity, while information about whether the phenomenon is characterized by replicative or stress-induced senescence, especially in non-aged organisms, is scarce. Herein, we subjected young mice to two different diet-induced NAFLD models which differed in the presence of obesity. In both models, liver fat accumulation and increased hepatic mRNA expression of steatosis-related genes were accompanied by hepatic senescence, indicated by the increased expression of senescence-associated genes and the presence of a robust hybrid histo-/immunochemical senescence-specific staining in the liver. Surprisingly, telomere length and global DNA methylation did not differ between the steatotic and the control livers, while malondialdehyde, a marker of oxidative stress, was upregulated in the mouse NAFLD livers. These findings suggest that senescence accompanies NAFLD emergence, even in non-aged organisms, and highlight the role of stress-induced senescence during steatosis development independently of obesity.
Collapse
Affiliation(s)
- Ioannis I. Moustakas
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.I.M.); (A.K.); (A.-I.L.); (A.-M.P.); (M.K.)
| | - Angeliki Katsarou
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.I.M.); (A.K.); (A.-I.L.); (A.-M.P.); (M.K.)
| | - Aigli-Ioanna Legaki
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.I.M.); (A.K.); (A.-I.L.); (A.-M.P.); (M.K.)
| | - Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (I.P.); (B.G.); (T.C.)
| | - Konstantinos Ntostoglou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (I.S.P.); (V.G.G.)
| | - Alkistis-Maria Papatheodoridi
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.I.M.); (A.K.); (A.-I.L.); (A.-M.P.); (M.K.)
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (I.P.); (B.G.); (T.C.)
| | - Ioannis S. Pateras
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (I.S.P.); (V.G.G.)
| | - Vassilis G. Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (I.S.P.); (V.G.G.)
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Molecular and Clinical Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4GJ, UK
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Michael Koutsilieris
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.I.M.); (A.K.); (A.-I.L.); (A.-M.P.); (M.K.)
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (I.P.); (B.G.); (T.C.)
| | - Antonios Chatzigeorgiou
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.I.M.); (A.K.); (A.-I.L.); (A.-M.P.); (M.K.)
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (I.P.); (B.G.); (T.C.)
| |
Collapse
|
31
|
Chen HJ, Li GL, Zhang WX, Fan J, Hu L, Zhang L, Zhang J, Yan YE. Maternal nicotine exposure during pregnancy and lactation induces brown adipose tissue whitening in female offspring. Toxicol Appl Pharmacol 2020; 409:115298. [PMID: 33091441 DOI: 10.1016/j.taap.2020.115298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/11/2020] [Accepted: 10/16/2020] [Indexed: 02/06/2023]
Abstract
Maternal nicotine exposure during pregnancy and lactation is associated with obesity in female offspring. Brown adipose tissue (BAT) is related to energy metabolism and obesity. In this study, we explored the mechanism of maternal nicotine exposure on BAT "whitening" in female offspring. Pregnant rats were randomly assigned to nicotine (1.0 mg/kg twice per day, subcutaneous administration) or control groups. The weight, structure, and microvascular density of interscapular BAT (iBAT) and the expression of PGC-1αUCP1 signals, mitochondrial biogenesis-related genes and angiogenesis-related genes were tested in 4- and 26-week-aged female offspring. In vitro, C3H10T1/2 cells were induced to differentiate into mature brown adipocytes, and 0-50 μM nicotine was treated on cells during the differentiation process. Nicotine-exposed females had higher iBAT weight, white-like adipocytes and abnormal mitochondrial structure in iBAT at 26 weeks rather than 4 weeks. The PGC-1αUCP1 signals and brown-like genes were down-regulated at 26 weeks, but the microvascular density and the expression of pro-angiogenic factors reduced more at 4 weeks in the nicotine group. In vitro, 50 μM nicotine significantly decreased the expression of PGC-1αUCP1 signals and angiogenesis-related genes. In conclusion, maternal nicotine exposure during pregnancy and lactation led to the "whitening" of BAT in adult female offspring: nicotine decreased BAT angiogenesis in the early development stage, and then, the impairment of blood vessels programed for the reduction of BAT phenotype through down-regulating the PGC-1αUCP1 signals in adulthood. This impairment of BAT may be a potential mechanism of nicotine-induced obesity in female offspring.
Collapse
Affiliation(s)
- Hui-Jian Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Gai-Ling Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Wan-Xia Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Jie Fan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Li Hu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Li Zhang
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Jing Zhang
- Center for Animal Experiment/Animal Biosafety Level 3 Laboratory, Wuhan University, Wuhan 430071, China
| | - You-E Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
| |
Collapse
|
32
|
Rodriguez-Ayala E, Gallegos-Cabrales EC, Gonzalez-Lopez L, Laviada-Molina HA, Salinas-Osornio RA, Nava-Gonzalez EJ, Leal-Berumen I, Escudero-Lourdes C, Escalante-Araiza F, Buenfil-Rello FA, Peschard VG, Laviada-Nagel A, Silva E, Veloz-Garza RA, Martinez-Hernandez A, Barajas-Olmos FM, Molina-Segui F, Gonzalez-Ramirez L, Espadas-Olivera R, Lopez-Muñoz R, Arjona-Villicaña RD, Hernandez-Escalante VM, Rodriguez-Arellano ME, Gaytan-Saucedo JF, Vaquera Z, Acebo-Martinez M, Cornejo-Barrera J, Jancy Andrea HQ, Castillo-Pineda JC, Murillo-Ramirez A, Diaz-Tena SP, Figueroa-Nuñez B, Valencia-Rendon ME, Garzon-Zamora R, Viveros-Paredes JM, Ángeles-Chimal J, Santa-Olalla Tapia J, Remes-Troche JM, Valdovinos-Chavez SB, Huerta-Avila EE, Lopez-Alvarenga JC, Comuzzie AG, Haack K, Han X, Orozco L, Weintraub S, Kent JW, Cole SA, Bastarrachea RA. Towards precision medicine: defining and characterizing adipose tissue dysfunction to identify early immunometabolic risk in symptom-free adults from the GEMM family study. Adipocyte 2020; 9:153-169. [PMID: 32272872 PMCID: PMC7153654 DOI: 10.1080/21623945.2020.1743116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/03/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022] Open
Abstract
Interactions between macrophages and adipocytes are early molecular factors influencing adipose tissue (AT) dysfunction, resulting in high leptin, low adiponectin circulating levels and low-grade metaflammation, leading to insulin resistance (IR) with increased cardiovascular risk. We report the characterization of AT dysfunction through measurements of the adiponectin/leptin ratio (ALR), the adipo-insulin resistance index (Adipo-IRi), fasting/postprandial (F/P) immunometabolic phenotyping and direct F/P differential gene expression in AT biopsies obtained from symptom-free adults from the GEMM family study. AT dysfunction was evaluated through associations of the ALR with F/P insulin-glucose axis, lipid-lipoprotein metabolism, and inflammatory markers. A relevant pattern of negative associations between decreased ALR and markers of systemic low-grade metaflammation, HOMA, and postprandial cardiovascular risk hyperinsulinemic, triglyceride and GLP-1 curves was found. We also analysed their plasma non-coding microRNAs and shotgun lipidomics profiles finding trends that may reflect a pattern of adipose tissue dysfunction in the fed and fasted state. Direct gene differential expression data showed initial patterns of AT molecular signatures of key immunometabolic genes involved in AT expansion, angiogenic remodelling and immune cell migration. These data reinforce the central, early role of AT dysfunction at the molecular and systemic level in the pathogenesis of IR and immunometabolic disorders.
Collapse
Affiliation(s)
- Ernesto Rodriguez-Ayala
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | | | - Laura Gonzalez-Lopez
- Dirección de Postgrado e Investigación, Universidad del Valle de Atemajac (UNIVA), Zapopan, México
| | | | - Rocio A. Salinas-Osornio
- Dirección de Postgrado e Investigación, Universidad del Valle de Atemajac (UNIVA), Zapopan, México
| | | | - Irene Leal-Berumen
- Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, México
| | | | - Fabiola Escalante-Araiza
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | - Fatima A. Buenfil-Rello
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Vanessa-Giselle Peschard
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | - Antonio Laviada-Nagel
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Eliud Silva
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | - Rosa A. Veloz-Garza
- Facultad de Enfermería, Universidad Autónoma de Nuevo León (UANL), Monterrey, México
| | - Angelica Martinez-Hernandez
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | - Francisco M. Barajas-Olmos
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | | | | | | | - Ricardo Lopez-Muñoz
- Escuela de Ciencias de la Salud, Universidad Marista de Mérida, Yucatán, Mexico
| | | | - Victor M. Hernandez-Escalante
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | | | - Janeth F. Gaytan-Saucedo
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Zoila Vaquera
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | | | - Judith Cornejo-Barrera
- Departamento de Enseñanza, Postgrado e Investigación, Hospital Infantil de Tamaulipas, Ciudad, México
| | - Huertas-Quintero Jancy Andrea
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | | | | | - Sara P. Diaz-Tena
- Departamento de Nutrición Humana, Universidad Latina de América, Morelia, México
| | | | | | - Rafael Garzon-Zamora
- Dirección de Postgrado e Investigación, Universidad del Valle de Atemajac (UNIVA), Zapopan, México
| | | | - José Ángeles-Chimal
- Facultad de Medicina, Universidad Autónoma Estado de Morelos, Cuernavaca, México
| | | | - José M. Remes-Troche
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, México
| | | | - Eira E. Huerta-Avila
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | - Juan Carlos Lopez-Alvarenga
- School of Medicine & South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | | | - Karin Haack
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Xianlin Han
- Department of Medicine, Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Lorena Orozco
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | - Susan Weintraub
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jack W. Kent
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Shelley A. Cole
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Raul A. Bastarrachea
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| |
Collapse
|
33
|
Hypoxia Pathway Proteins are Master Regulators of Erythropoiesis. Int J Mol Sci 2020; 21:ijms21218131. [PMID: 33143240 PMCID: PMC7662373 DOI: 10.3390/ijms21218131] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/21/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Erythropoiesis is a complex process driving the production of red blood cells. During homeostasis, adult erythropoiesis takes place in the bone marrow and is tightly controlled by erythropoietin (EPO), a central hormone mainly produced in renal EPO-producing cells. The expression of EPO is strictly regulated by local changes in oxygen partial pressure (pO2) as under-deprived oxygen (hypoxia); the transcription factor hypoxia-inducible factor-2 induces EPO. However, erythropoiesis regulation extends beyond the well-established hypoxia-inducible factor (HIF)-EPO axis and involves processes modulated by other hypoxia pathway proteins (HPPs), including proteins involved in iron metabolism. The importance of a number of these factors is evident as their altered expression has been associated with various anemia-related disorders, including chronic kidney disease. Eventually, our emerging understanding of HPPs and their regulatory feedback will be instrumental in developing specific therapies for anemic patients and beyond.
Collapse
|
34
|
Kochumon S, Al Madhoun A, Al-Rashed F, Thomas R, Sindhu S, Al-Ozairi E, Al-Mulla F, Ahmad R. Elevated adipose tissue associated IL-2 expression in obesity correlates with metabolic inflammation and insulin resistance. Sci Rep 2020; 10:16364. [PMID: 33004937 PMCID: PMC7530670 DOI: 10.1038/s41598-020-73347-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/16/2020] [Indexed: 12/11/2022] Open
Abstract
Adipose tissue (AT) associated cytokines are involved in the development of chronic low-grade inflammation in obese individuals. IL-2, a pleiotropic cytokine, contributes to immune alterations during inflammation. However, the interaction between AT-IL-2 and other inflammatory biomolecules in obesity remains elusive. We investigated whether AT-IL-2 expression was associated with markers of inflammation and insulin resistance in overweight/obese individuals. Subcutaneous fat tissues were collected from 56 individuals (lean/overweight/obese) for RNA extraction. IL-2 and inflammatory mediators were quantified by qRT-PCR and immunohistochemistry. CRP was measured by ELISA. AT-IL-2 expression was higher in obese compared with lean individuals (P < 0.021) and correlated with BMI. IL-2 correlated with interleukins IL-8 and IL-12A (r = 0.333-0.481; p = 0.0001-0.029); as well as with chemokines and their receptors including CCL5, CCL19, CCR2 and CCR5 (r = 0.538-0.677; p < 0.0001). Moreover, IL-2 correlated with toll-like receptors (TLR2, TLR8, TLR10), interferon regulatory factor 5 (IRF5) and cluster of differentiation CD11c (r = 0.282-0.357; p < 0.039). Notably, IL-2 was associated positively with fasting blood glucose (FBG), HbA1c, TGL and CRP (r ≥ 0.423;P ≤ 0.007). In multiple regression analysis, IL-2 is an independent predictor of IL-8, IL-12A, TLR10, TGL and HbA1c. Overall, our data demonstrate that increased expression of the AT-IL-2, in obesity, may represent a novel biomarker for progression of metabolic inflammation and insulin-resistance.
Collapse
Affiliation(s)
- Shihab Kochumon
- Immunology and Microbiology Department, Dasman Diabetes Institute, Jasim Mohamad Al Bahar St., P.O. Box 1180, 15462, Kuwait City, Kuwait
| | - Ashraf Al Madhoun
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Kuwait City, Kuwait
- Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fatema Al-Rashed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Jasim Mohamad Al Bahar St., P.O. Box 1180, 15462, Kuwait City, Kuwait
- Immunology Department, Ministry of Health, Kuwait City, Kuwait
| | - Reeby Thomas
- Immunology and Microbiology Department, Dasman Diabetes Institute, Jasim Mohamad Al Bahar St., P.O. Box 1180, 15462, Kuwait City, Kuwait
| | - Sardar Sindhu
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Ebaa Al-Ozairi
- Medical Division, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Jasim Mohamad Al Bahar St., P.O. Box 1180, 15462, Kuwait City, Kuwait.
| |
Collapse
|
35
|
Pyrina I, Chung KJ, Michailidou Z, Koutsilieris M, Chavakis T, Chatzigeorgiou A. Fate of Adipose Progenitor Cells in Obesity-Related Chronic Inflammation. Front Cell Dev Biol 2020; 8:644. [PMID: 32760729 PMCID: PMC7372115 DOI: 10.3389/fcell.2020.00644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/26/2020] [Indexed: 12/16/2022] Open
Abstract
Adipose progenitor cells, or preadipocytes, constitute a small population of immature cells within the adipose tissue. They are a heterogeneous group of cells, in which different subtypes have a varying degree of commitment toward diverse cell fates, contributing to white and beige adipogenesis, fibrosis or maintenance of an immature cell phenotype with proliferation capacity. Mature adipocytes as well as cells of the immune system residing in the adipose tissue can modulate the function and differentiation potential of preadipocytes in a contact- and/or paracrine-dependent manner. In the course of obesity, the accumulation of immune cells within the adipose tissue contributes to the development of a pro-inflammatory microenvironment in the tissue. Under such circumstances, the crosstalk between preadipocytes and immune or parenchymal cells of the adipose tissue may critically regulate the differentiation of preadipocytes into white adipocytes, beige adipocytes, or myofibroblasts, thereby influencing adipose tissue expansion and adipose tissue dysfunction, including downregulation of beige adipogenesis and development of fibrosis. The present review will outline the current knowledge about factors shaping cell fate decisions of adipose progenitor cells in the context of obesity-related inflammation.
Collapse
Affiliation(s)
- Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Zoi Michailidou
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Antonios Chatzigeorgiou
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
36
|
Troullinaki M, Chen LS, Witt A, Pyrina I, Phieler J, Kourtzelis I, Chmelar J, Sprott D, Gercken B, Koutsilieris M, Chavakis T, Chatzigeorgiou A. Robo4-mediated pancreatic endothelial integrity decreases inflammation and islet destruction in autoimmune diabetes. FASEB J 2020; 34:3336-3346. [PMID: 31916652 DOI: 10.1096/fj.201900125rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/18/2022]
Abstract
In Type 1 Diabetes Mellitus (T1DM), leukocyte infiltration of the pancreatic islets and the resulting immune-mediated destruction of beta cells precede hyperglycemia and clinical disease symptoms. In this context, the role of the pancreatic endothelium as a barrier for autoimmunity- and inflammation-related destruction of the islets is not well studied. Here, we identified Robo4, expressed on endothelial cells, as a regulator of pancreatic vascular endothelial permeability during autoimmune diabetes. Circulating levels of Robo4 were upregulated in mice subjected to the Multiple Low-Dose Streptozotocin (MLDS) model of diabetes. Upon MLDS induction, Robo4-deficiency resulted in increased pancreatic vascular permeability, leukocyte infiltration to the islets and islet apoptosis, associated with reduced insulin levels and faster diabetes development. On the contrary, in vivo administration of Slit2 in mice modestly delayed the emergence of hyperglycaemia and ameliorated islet inflammation in MLDS-induced diabetes. Thus, Robo4-mediated endothelial barrier integrity reduces insulitis and islet destruction in autoimmune diabetes. Our findings highlight the importance of the endothelium as gatekeeper of pancreatic inflammation during T1DM development and may pave the way for novel Robo4-related therapeutic approaches for autoimmune diabetes.
Collapse
Affiliation(s)
- Maria Troullinaki
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Lan-Sun Chen
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anke Witt
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Julia Phieler
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ioannis Kourtzelis
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jindrich Chmelar
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - David Sprott
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Antonios Chatzigeorgiou
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
37
|
Kochumon S, Madhoun AA, Al-Rashed F, Azim R, Al-Ozairi E, Al-Mulla F, Ahmad R. Adipose tissue gene expression of CXCL10 and CXCL11 modulates inflammatory markers in obesity: implications for metabolic inflammation and insulin resistance. Ther Adv Endocrinol Metab 2020; 11:2042018820930902. [PMID: 32655851 PMCID: PMC7331767 DOI: 10.1177/2042018820930902] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 05/10/2020] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The CXCL subfamily of chemokines (CXCL9, CXCL10, and CXCL11; angiostatic chemokines) plays a key role in many inflammatory diseases. However, the expression of CXCLs in adipose tissue (AT) during obesity and association of these CXCLs with inflammatory markers and insulin resistance are poorly understood. Therefore, this study aimed to investigate the effects of CXCL gene expression on subcutaneous AT inflammatory markers and insulin resistance. METHODS Subcutaneous-fat biopsies were collected from 59 nondiabetic (lean/overweight/obese) individuals for RNA isolation. Expression levels of AT CXCL and inflammatory markers were determined by quantitative reverse transcriptase polymerase chain reaction (RT-qPCR). Biomedical parameters in the plasma were measured by enzyme-linked immunosorbent assay (ELISA). Insulin resistance was estimated using homeostatic model assessment (HOMA-IR). RESULTS AT CXCL expression was higher in obese compared with lean individuals (p < 0.05) and positively correlated with body mass index (BMI; r ⩾ 0.269, p < 0.05). Expression of CXCL9, CXCL10, and CXCL11 correlated significantly with various pro-inflammatory markers, including family members of interleukins, chemokines, and their prospective receptors (r ⩾ 0.339, p ⩽ 0.009), but not anti-inflammatory markers. CXCL11 expression correlated specifically with the expression of CCL5, CCL18, TLR3, TLR4, TLR8, IRF5, and NF-κB (r ⩾ 0.279, p ⩽ 0.039). Notably, CXCL11 was correlated with C-reactive protein (CRP), fasting blood glucose (FBG), and HOMA-IR. In multiple regression analysis, CXCL11 was identified as an independent predictor of CCL19, CCL5, IL-6, and TLR3. CONCLUSION These data suggest that the CXCL family members, specifically CXCL10 and CXCL11, are potential biomarkers for the onset of AT inflammation during obesity.
Collapse
Affiliation(s)
| | | | - Fatema Al-Rashed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Rafaat Azim
- School of Medicine, Royal College of Surgeons in Ireland, Medical University of Bahrain, Busaiteen, Bahrain
| | | | - Fahd Al-Mulla
- Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait, Dasman, Kuwait
| | | |
Collapse
|
38
|
Michailidou Z. Fundamental roles for hypoxia signalling in adipose tissue metabolism and inflammation in obesity. CURRENT OPINION IN PHYSIOLOGY 2019. [DOI: 10.1016/j.cophys.2019.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
39
|
Adipocyte Hypoxia-Inducible Factor 2α Suppresses Atherosclerosis by Promoting Adipose Ceramide Catabolism. Cell Metab 2019; 30:937-951.e5. [PMID: 31668872 DOI: 10.1016/j.cmet.2019.09.016] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/10/2019] [Accepted: 09/26/2019] [Indexed: 12/18/2022]
Abstract
Obesity-induced adipose dysfunction is a major contributor to atherosclerosis. Cold exposure has been reported to affect atherosclerosis through regulation of adipose function, but the mechanism has not been well clarified. Here, adipocyte hypoxia-inducible factor 2α (HIF-2α) was upregulated after mild cold exposure at 16°C and mediated cold-induced thermogenesis. Adipocyte HIF-2α deficiency exacerbated Western-diet-induced atherosclerosis by increasing adipose ceramide levels, which blunted hepatocyte cholesterol elimination and thermogenesis. Mechanistically, Acer2, the gene encoding alkaline ceramidase 2, was identified as a novel target gene of HIF-2α, triggering ceramide catabolism. Adipose overexpression of ACER2 rescued adipocyte HIF-2α-deficiency-induced exacerbation of atherosclerosis. Furthermore, activation of adipose HIF-2α by the HIF prolyl hydroxylase inhibitor FG-4592 had protective effects on atherosclerosis, accompanied by a reduction in adipose and plasma ceramide and plasma cholesterol levels. This study highlights adipocyte HIF-2α as a putative drug target against atherosclerosis.
Collapse
|
40
|
Fan L, Ye H, Wan Y, Qin L, Zhu L, Su J, Zhu X, Zhang L, Miao Q, Zhang Q, Zhang Z, Xu A, Li Y, Li X, Wang Y. Adaptor protein APPL1 coordinates HDAC3 to modulate brown adipose tissue thermogenesis in mice. Metabolism 2019; 100:153955. [PMID: 31390528 DOI: 10.1016/j.metabol.2019.153955] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/12/2019] [Accepted: 07/31/2019] [Indexed: 01/17/2023]
Abstract
OBJECTIVES The activation of brown adipose tissue (BAT) is considered as a promising therapeutic target for obesity. APPL1 (Adaptor protein containing the Pleckstrin homology domain, Phosphotyrosine binding domain and Leucine zipper motif) is an intracellular adaptor protein and its genetic variation is correlated with BMI and body fat distribution in diabetic patients. However, little is known about the roles of APPL1 in BAT thermogenesis. MATERIALS/METHODS In this study, adipose tissue specific knockout (ASKO) mice were generated to evaluate APPL1's role in BAT thermogenesis in vivo, and possible signaling pathways were further explored in cultured brown adipocytes. RESULTS After high fat diet challenge, APPL1 ASKO mice developed more severe obesity, glucose intolerance and insulin resistance compared with control mice. Metabolic cage study showed that APPL1 deficiency impaired energy expenditure and adaptive thermogenesis in ASKO mice. PET-CT analysis showed decreased standardized uptake value (SUV) in the inter-scapular region which indicated impaired BAT activity in ASKO mice. Further study showed deletion of APPL1 attenuated brown fat specific gene expression, such as UCP1 and PGC1α in both BAT and brown adipocytes. In cultured brown adipocytes, upon cAMP stimulation, APPL1 shuttled from cytosol to nuclei. Co-IP and ChIP study showed that APPL1 could directly interact with histone deacetylase 3 (HDAC3) to mediate chromatin remodeling and UCP1 gene expression. CONCLUSIONS Our data demonstrated the essential role of APPL1 in regulating brown adipocytes thermogenesis via interaction with HDAC3, which may have potential therapeutic implications for treatment of obesity.
Collapse
Affiliation(s)
- Linling Fan
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongying Ye
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yun Wan
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Lang Qin
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Lu Zhu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Su
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoming Zhu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Lv Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Miao
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiongyue Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhaoyun Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Aimin Xu
- Department of Medicine, the University of Hong Kong, Hong Kong
| | - Yiming Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Xi Li
- Institute of Life Sciences, Chongqing Medical University, China.
| | - Yi Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
41
|
Feng Z, Wei Y, Zhang Y, Qiu Y, Liu X, Su L, Liang N, Yin H, Ding Q. Identification of a rhodanine derivative BML-260 as a potent stimulator of UCP1 expression. Theranostics 2019; 9:3501-3514. [PMID: 31281493 PMCID: PMC6587176 DOI: 10.7150/thno.31951] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 04/14/2019] [Indexed: 01/23/2023] Open
Abstract
Identification of proper agents to increase or activate UCP1+ cells in adipose tissues remains a potent therapeutic strategy to combat obesity. Screening systems for UCP1 activators have been previously established and allow for unbiased discovery of effective compound(s). Methods: A previously established Ucp1-2A-GFP reporter system was applied to a chemical library containing 33 phosphatase inhibitors. Compounds that can significantly activate UCP1 expression were further tested in vivo in mouse adipose tissues. Possible underlying mechanism was explored via RNA profiling, CMAP analysis, CRISPR targeting as well as inhibitor treatments. Results: We identified BML-260, a known potent inhibitor of the dual-specific phosphatase JSP-1, that significantly increased UCP1 expression in both brown and white adipocytes. BML-260 treatment also activated oxidative phosphorylation genes, increased mitochondrial activity as well as heat generation in vitro and in vivo. Mechanistic studies revealed that effect of BML-260 on adipocytes was partly through activated CREB, STAT3 and PPAR signaling pathways, and was unexpectedly JSP-1 independent. Conclusion: The rhodanine derivate BML-260 was previously identified to be a JSP-1 inhibitor, and thus was proposed to treat inflammatory and proliferative disorders associated with dysfunctional JNK signaling. This work provides evidences that BML-260 can also exert a JSP-1-independent effect in activating UCP1 and thermogenesis in adipocytes, and be potentially applied to treat obesity.
Collapse
|
42
|
Wielockx B, Grinenko T, Mirtschink P, Chavakis T. Hypoxia Pathway Proteins in Normal and Malignant Hematopoiesis. Cells 2019; 8:cells8020155. [PMID: 30781787 PMCID: PMC6406588 DOI: 10.3390/cells8020155] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/25/2022] Open
Abstract
The regulation of oxygen (O₂) levels is crucial in embryogenesis and adult life, as O₂ controls a multitude of key cellular functions. Low oxygen levels (hypoxia) are relevant for tissue physiology as they are integral to adequate metabolism regulation and cell fate. Hence, the hypoxia response is of utmost importance for cell, organ and organism function and is dependent on the hypoxia-inducible factor (HIF) pathway. HIF pathway activity is strictly regulated by the family of oxygen-sensitive HIF prolyl hydroxylase domain (PHD) proteins. Physiologic hypoxia is a hallmark of the hematopoietic stem cell (HSC) niche in the bone marrow. This niche facilitates HSC quiescence and survival. The present review focuses on current knowledge and the many open questions regarding the impact of PHDs/HIFs and other proteins of the hypoxia pathway on the HSC niche and on normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Ben Wielockx
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany.
| | - Tatyana Grinenko
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany.
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany.
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany.
| |
Collapse
|
43
|
Grzybek M, Palladini A, Alexaki VI, Surma MA, Simons K, Chavakis T, Klose C, Coskun Ü. Comprehensive and quantitative analysis of white and brown adipose tissue by shotgun lipidomics. Mol Metab 2019; 22:12-20. [PMID: 30777728 PMCID: PMC6437637 DOI: 10.1016/j.molmet.2019.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/17/2019] [Accepted: 01/23/2019] [Indexed: 12/12/2022] Open
Abstract
Objective Shotgun lipidomics enables an extensive analysis of lipids from tissues and fluids. Each specimen requires appropriate extraction and processing procedures to ensure good coverage and reproducible quantification of the lipidome. Adipose tissue (AT) has become a research focus with regard to its involvement in obesity-related pathologies. However, the quantification of the AT lipidome is particularly challenging due to the predominance of triacylglycerides, which elicit high ion suppression of the remaining lipid classes. Methods We present a new and validated method for shotgun lipidomics of AT, which tailors the lipid extraction procedure to the target specimen and features high reproducibility with a linear dynamic range of at least 4 orders of magnitude for all lipid classes. Results Utilizing this method, we observed tissue-specific and diet-related differences in three AT types (brown, gonadal, inguinal subcutaneous) from lean and obese mice. Brown AT exhibited a distinct lipidomic profile with the greatest lipid class diversity and responded to high-fat diet by altering its lipid composition, which shifted towards that of white AT. Moreover, diet-induced obesity promoted an overall remodeling of the lipidome, where all three AT types featured a significant increase in longer and more unsaturated triacylglyceride and phospholipid species. Conclusions The here presented method facilitates reproducible systematic lipidomic profiling of AT and could be integrated with further –omics approaches used in (pre-) clinical research, in order to advance the understanding of the molecular metabolic dynamics involved in the pathogenesis of obesity-associated disorders. Validated shotgun lipidomics method of AT covering 300 lipids of 20 classes and linear dynamic range of 4 orders of magnitude. Increase of longer and more unsaturated triacylglycerides and phospholipids in brown and white AT under high-fat diet. Differences in the lipidomes of gonadal, subcutaneous and brown AT.
Collapse
Affiliation(s)
- Michal Grzybek
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Alessandra Palladini
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Vasileia I Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, Dresden, Germany
| | | | | | - Triantafyllos Chavakis
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, Dresden, Germany
| | | | - Ünal Coskun
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.
| |
Collapse
|
44
|
Abstract
Hypoxia-inducible factors (HIFs), a family of transcription factors activated by hypoxia, consist of three α-subunits (HIF1α, HIF2α and HIF3α) and one β-subunit (HIF1β), which serves as a heterodimerization partner of the HIFα subunits. HIFα subunits are stabilized from constitutive degradation by hypoxia largely through lowering the activity of the oxygen-dependent prolyl hydroxylases that hydroxylate HIFα, leading to their proteolysis. HIF1α and HIF2α are expressed in different tissues and regulate target genes involved in angiogenesis, cell proliferation and inflammation, and their expression is associated with different disease states. HIFs have been widely studied because of their involvement in cancer, and HIF2α-specific inhibitors are being investigated in clinical trials for the treatment of kidney cancer. Although cancer has been the major focus of research on HIF, evidence has emerged that this pathway has a major role in the control of metabolism and influences metabolic diseases such as obesity, type 2 diabetes mellitus and non-alcoholic fatty liver disease. Notably increased HIF1α and HIF2α signalling in adipose tissue and small intestine, respectively, promotes metabolic diseases in diet-induced disease models. Inhibition of HIF1α and HIF2α decreases the adverse diet-induced metabolic phenotypes, suggesting that they could be drug targets for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.
| |
Collapse
|
45
|
Gaspar JM, Velloso LA. Hypoxia Inducible Factor as a Central Regulator of Metabolism - Implications for the Development of Obesity. Front Neurosci 2018; 12:813. [PMID: 30443205 PMCID: PMC6221908 DOI: 10.3389/fnins.2018.00813] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/18/2018] [Indexed: 12/19/2022] Open
Abstract
The hypothalamus plays a major role in the regulation of food intake and energy expenditure. In the last decade, it was demonstrated that consumption of high-fat diets triggers the activation of an inflammatory process in the hypothalamus, inducing neurofunctional alterations and contributing to the development of obesity. Hypoxia-inducible factors (HIFs) are key molecules that regulate cellular responses to inflammation and hypoxia, being essential for the normal cell function and survival. Currently, evidence points to a role of HIF pathway in metabolic regulation that could also be involved in the progression of obesity and metabolic diseases. The challenge is to understand how HIF modulation impacts body mass gain and metabolic disorders such as insulin resistance. Distinct animal models with tissue-specific knocking-out or overexpression of hypoxia signaling pathway genes revealed a cell-specificity in the activation of HIF pathways, and some of them have opposite phenotypes among the various HIFs gain- and loss-of-function mouse models. In this review, we discuss the major findings that provide support for a role of HIF pathway involvement in the regulation of metabolism, especially in glucose and energy homeostasis.
Collapse
Affiliation(s)
- Joana M Gaspar
- Post-Graduation in Biochemistry, Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Brazil.,Laboratório de Bioenergética e Estresse Oxidativo, Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Lício A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, Brazil
| |
Collapse
|
46
|
Qiu Y, Sun Y, Xu D, Yang Y, Liu X, Wei Y, Chen Y, Feng Z, Li S, Reyad-Ul Ferdous M, Zhao Y, Xu H, Lao Y, Ding Q. Screening of FDA-approved drugs identifies sutent as a modulator of UCP1 expression in brown adipose tissue. EBioMedicine 2018; 37:344-355. [PMID: 30348622 PMCID: PMC6286640 DOI: 10.1016/j.ebiom.2018.10.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The pharmacological activation of thermogenesis in brown adipose tissue has long been considered promising strategies to treat obesity. However, identification of safe and effective agents remains a challenge. In this study, we addressed this challenge by developing a cellular system with a fluorescence readout, and applied in a high-throughput manner to screen for FDA-approved drugs that may activate endogenous UCP1 expression in adipocytes. METHODS We have generated a Ucp1-2A-GFP reporter mouse, in which GFP intensity serves as a surrogate of the endogenous expression level of UCP1 protein; and immortalized brown adipocytes were derived from this mouse model and applied in drug screening. Candidate drugs were further tested in mouse models either fed with normal chow or high fat diet to induce obesity. FINDINGS By using the cellular screening platform, we identified a group of FDA-approved drugs that can upregulate UCP1 expression in brown adipocyte, including previously known UCP1 activators and new candidate drugs. Further studies focusing on a previously unreported drug-sutent, revealed that sutent treatment could increase the energy expenditure and inhibit lipid synthesis in mouse adipose and liver tissues, resulting in improved metabolism and resistance to obesity. INTERPRETATION This study offered an easy-to-use cellular screening system for UCP1 activators, and provided a candidate list of FDA-approved drugs that can potentially treat obesity. Further study of these candidates may shed new light on the drug discovery towards obesity. FUND: National Key Research and Development Program and the Strategic Priority Research Program of the Chinese Academy of Sciences, etc. (250 words).
Collapse
Affiliation(s)
- Yan Qiu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Yingmin Sun
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Danqing Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Yuanyuan Yang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Xiaojian Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Yuda Wei
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Yanhao Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Zhuanghui Feng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Shuang Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Md Reyad-Ul Ferdous
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Yongxu Zhao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, PR China
| | - Yuanzhi Lao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, PR China.
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, PR China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, PR China.
| |
Collapse
|
47
|
Gaspar JM, Mendes NF, Corrêa-da-Silva F, Lima-Junior JCD, Gaspar RC, Ropelle ER, Araujo EP, Carvalho HM, Velloso LA. Downregulation of HIF complex in the hypothalamus exacerbates diet-induced obesity. Brain Behav Immun 2018; 73:550-561. [PMID: 29935943 DOI: 10.1016/j.bbi.2018.06.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/30/2018] [Accepted: 06/20/2018] [Indexed: 11/25/2022] Open
Abstract
Hypothalamic hypoxia-inducible factor-1 (HIF-1) can regulate whole-body energy homeostasis in response to changes in blood glucose, suggesting that it acts as a sensor for systemic energy stores. Here, we hypothesized that hypothalamic HIF-1 could be affected by diet-induced obesity (DIO). We used eight-week old, male C57Bl6 mice, fed normal chow diet or with high fat diet for 1, 3, 7, 14 and 28 days. The expression of HIF-1alpha and HIF-1beta was measured by PCR and western blotting and its hypothalamic distribution was evaluated by fluorescence microscopy. Inhibition of HIF-1beta in arcuate nucleus of hypothalamus was performed using stereotaxic injection of shRNA lentiviral particles and animals were grouped under normal chow diet or high fat diet for 14 days. Using bioinformatics, we show that in humans, the levels of HIF-1 transcripts are directly correlated with those of hypothalamic transcripts for proteins involved in inflammation, regulation of apoptosis, autophagy, and the ubiquitin/proteasome system; furthermore, in rodents, hypothalamic HIF-1 expression is directly correlated with the phenotype of increased energy expenditure. In mice, DIO was accompanied by increased HIF-1 expression. The inhibition of hypothalamic HIF-1 by injection of an shRNA resulted in a further increase in body mass, a decreased basal metabolic rate, increased hypothalamic inflammation, and glucose intolerance. Thus, hypothalamic HIF-1 is increased during DIO, and its inhibition worsens the obesity-associated metabolic phenotype. Thus, hypothalamic HIF-1 emerges as a target for therapeutic intervention against obesity.
Collapse
Affiliation(s)
- Joana M Gaspar
- Laboratory of Cell Signaling, University of Campinas, Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Natália Ferreira Mendes
- Laboratory of Cell Signaling, University of Campinas, Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil; Faculty of Nursing, University of Campinas, Campinas, São Paulo, Brazil
| | - Felipe Corrêa-da-Silva
- Laboratory of Cell Signaling, University of Campinas, Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - José C de Lima-Junior
- Laboratory of Cell Signaling, University of Campinas, Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Rodrigo C Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eliana P Araujo
- Laboratory of Cell Signaling, University of Campinas, Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil; Faculty of Nursing, University of Campinas, Campinas, São Paulo, Brazil
| | - Humberto M Carvalho
- Department of Physical Education, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Lício A Velloso
- Laboratory of Cell Signaling, University of Campinas, Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil.
| |
Collapse
|
48
|
Hueso L, Ortega R, Selles F, Wu-Xiong NY, Ortega J, Civera M, Ascaso JF, Sanz MJ, Real JT, Piqueras L. Upregulation of angiostatic chemokines IP-10/CXCL10 and I-TAC/CXCL11 in human obesity and their implication for adipose tissue angiogenesis. Int J Obes (Lond) 2018; 42:1406-1417. [PMID: 29795466 DOI: 10.1038/s41366-018-0102-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND/AIMS Impaired angiogenesis is linked to adipose tissue (AT) dysfunction, inflammation, and insulin resistance in human obesity. Chemokine (C-X-C motif) receptor. (CXCR3) ligands are important regulators of angiogenesis in different disease contexts such as cancer; however, their role in human morbid obesity is unknown. We investigated the role of the CXCR3 axis in AT angiogenesis in morbidly obese patients. SUBJECTS/METHODS The study group comprised 50 morbidly obese patients (mean age 44 ± 1 years, body mass index 44 ± 1 kg/m2) who had undergone laparoscopic Roux-Y-gastric bypass surgery, and 25 age-matched non-obese control subjects. We measured the circulating levels of the CXCR3 ligands monokine induced by interferon-γ (MIG/CXCL9), interferon-γ inducible protein 10 (IP-10/CXCL10), and interferon-γ-inducible T-cell alpha chemoattractant (I-TAC/CXCL11) in all studied subjects. Additionally, the expression of CXCR3 ligands was analyzed in paired biopsies of subcutaneous and visceral AT obtained during the laparoscopic procedure in morbidly obese patients. Additionally, we explored the functional role of CXCR3 ligands on angiogenesis in AT from morbidly obese patients using an ex vivo assay. RESULTS Plasma levels of CXCL10 and CXCL11 were significantly higher in morbidly obese patients than in controls (p < 0.01). In ex vivo assays, angiogenic growth was markedly lower in visceral AT than in subcutaneous AT (p < 0.05), which was related to significant tissue upregulation of CXCL10, CXCL11 and CXCR3 (p < 0.05). CXCL10 or CXCL11 inhibited AT angiogenesis (p < 0.05), and blockade of CXCR3 function significantly increased capillary sprouting in visceral fat deposits (p < 0.05). Western blot analysis showed that the p38 mitogen-activated protein kinase signaling pathway was implicated in the angiostatic effects of CXCR3 in AT. CONCLUSIONS CXCL10 and CXCL11 may play. deleterious role in obesity as potential inhibitors of AT angiogenesis. Accordingly, pharmacological blockade of CXCR3 could represent. therapy to prevent AT dysfunction in obesity.
Collapse
Affiliation(s)
- Luisa Hueso
- Institute of Health Research-INCLIVA, Valencia, Spain
| | - Rebeca Ortega
- Institute of Health Research-INCLIVA, Valencia, Spain.,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | | | - Ning Yun Wu-Xiong
- Institute of Health Research-INCLIVA, Valencia, Spain.,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain
| | - Joaquin Ortega
- Surgery Service, University Clinic Hospital of Valencia, Valencia, Spain.,Department of Surgery, University of Valencia, Valencia, Spain
| | - Miguel Civera
- Institute of Health Research-INCLIVA, Valencia, Spain.,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain
| | - Juan F Ascaso
- Institute of Health Research-INCLIVA, Valencia, Spain.,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain.,CIBERDEM: Diabetes and Associated Metabolic Diseases Networking Biomedical Research- ISCIII, Madrid, Spain
| | - Maria-Jesus Sanz
- Institute of Health Research-INCLIVA, Valencia, Spain.,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - José T Real
- Institute of Health Research-INCLIVA, Valencia, Spain. .,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain. .,CIBERDEM: Diabetes and Associated Metabolic Diseases Networking Biomedical Research- ISCIII, Madrid, Spain.
| | - Laura Piqueras
- Institute of Health Research-INCLIVA, Valencia, Spain. .,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain.
| |
Collapse
|
49
|
Kumari M, Heeren J, Scheja L. Regulation of immunometabolism in adipose tissue. Semin Immunopathol 2017; 40:189-202. [PMID: 29209828 DOI: 10.1007/s00281-017-0668-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 11/22/2017] [Indexed: 12/14/2022]
|
50
|
Alexaki VI, May AE, Fujii C, Mund C, Gawaz M, Ungern-Sternberg SNIV, Chavakis T, Seizer P. S100A9 induces monocyte/ macrophage migration via EMMPRIN. Thromb Haemost 2017; 117:636-639. [DOI: 10.1160/th16-06-0434] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/16/2016] [Indexed: 01/26/2023]
|