1
|
Müller L, Hoffmann A, Bernhart SH, Ghosh A, Zhong J, Hagemann T, Sun W, Dong H, Noé F, Wolfrum C, Dietrich A, Stumvoll M, Massier L, Blüher M, Kovacs P, Chakaroun R, Keller M. Blood methylation pattern reflects epigenetic remodelling in adipose tissue after bariatric surgery. EBioMedicine 2024; 106:105242. [PMID: 39002385 PMCID: PMC11284569 DOI: 10.1016/j.ebiom.2024.105242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND Studies on DNA methylation following bariatric surgery have primarily focused on blood cells, while it is unclear to which extend it may reflect DNA methylation profiles in specific metabolically relevant organs such as adipose tissue. Here, we investigated whether adipose tissue depots specific methylation changes after bariatric surgery are mirrored in blood. METHODS Using Illumina 850K EPIC technology, we analysed genome-wide DNA methylation in paired blood, subcutaneous and omental visceral AT (SAT/OVAT) samples from nine individuals (N = 6 female) with severe obesity pre- and post-surgery. FINDINGS The numbers and effect sizes of differentially methylated regions (DMRs) post-bariatric surgery were more pronounced in AT (SAT: 12,865 DMRs from -11.5 to 10.8%; OVAT: 14,632 DMRs from -13.7 to 12.8%) than in blood (9267 DMRs from -8.8 to 7.7%). Cross-tissue DMRs implicated immune-related genes. Among them, 49 regions could be validated with similar methylation changes in blood from independent individuals. Fourteen DMRs correlated with differentially expressed genes in AT post bariatric surgery, including downregulation of PIK3AP1 in both SAT and OVAT. DNA methylation age acceleration was significantly higher in AT compared to blood, but remained unaffected after surgery. INTERPRETATION Concurrent methylation pattern changes in blood and AT, particularly in immune-related genes, suggest blood DNA methylation mirrors AT's inflammatory state post-bariatric surgery. FUNDING The funding sources are listed in the Acknowledgments section.
Collapse
Affiliation(s)
- Luise Müller
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany
| | - Stephan H Bernhart
- Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107, Leipzig, Germany; Bioinformatics Group, Department of Computer, University of Leipzig, 04107, Leipzig, Germany; Transcriptome Bioinformatics, LIFE Research Center for Civilization Diseases, University of Leipzig, 04107, Leipzig, Germany
| | - Adhideb Ghosh
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Jiawei Zhong
- Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, 141 83, Huddinge, Sweden
| | - Tobias Hagemann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany
| | - Wenfei Sun
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Hua Dong
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Falko Noé
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Arne Dietrich
- Leipzig University Hospital, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Section of Bariatric Surgery, 04103, Leipzig, Germany
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany; Deutsches Zentrum für Diabetesforschung e.V., 85764, Neuherberg, Germany
| | - Lucas Massier
- Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, 141 83, Huddinge, Sweden
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany; Deutsches Zentrum für Diabetesforschung e.V., 85764, Neuherberg, Germany
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany; Deutsches Zentrum für Diabetesforschung e.V., 85764, Neuherberg, Germany
| | - Rima Chakaroun
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany; The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 41345, Gothenburg, Sweden
| | - Maria Keller
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, 04103, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany.
| |
Collapse
|
2
|
Morris I, Croes CA, Boes M, Kalkhoven E. Advanced omics techniques shed light on CD1d-mediated lipid antigen presentation to iNKT cells. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159292. [PMID: 36773690 DOI: 10.1016/j.bbalip.2023.159292] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/26/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Invariant natural killer T cells (iNKT cells) can be activated through binding antigenic lipid/CD1d complexes to their TCR. Antigenic lipids are processed, loaded, and displayed in complex with CD1d by lipid antigen presenting cells (LAPCs). The mechanism of lipid antigen presentation via CD1d is highly conserved with recent work showing adipocytes are LAPCs that, besides having a role in lipid storage, can activate iNKT cells and play an important role in systemic metabolic disease. Recent studies shed light on parameters potentially dictating cytokine output and how obesity-associated metabolic disease may affect such parameters. By following a lipid antigen's journey, we identify five key areas which may dictate cytokine skew: co-stimulation, structural properties of the lipid antigen, stability of lipid antigen/CD1d complexes, intracellular and extracellular pH, and intracellular and extracellular lipid environment. Recent publications indicate that the combination of advanced omics-type approaches and machine learning may be a fruitful way to interconnect these 5 areas, with the ultimate goal to provide new insights for therapeutic exploration.
Collapse
Affiliation(s)
- Imogen Morris
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584, CG, Utrecht, the Netherlands
| | - Cresci-Anne Croes
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, 6708WE Wageningen, the Netherlands
| | - Marianne Boes
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584, EA, Utrecht, the Netherlands; Department of Paediatric Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584, EA, Utrecht, the Netherlands
| | - Eric Kalkhoven
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584, CG, Utrecht, the Netherlands.
| |
Collapse
|
3
|
Subramanian N, Hofwimmer K, Tavira B, Massier L, Andersson DP, Arner P, Laurencikiene J. Adipose tissue specific CCL18 associates with cardiometabolic diseases in non-obese individuals implicating CD4 + T cells. Cardiovasc Diabetol 2023; 22:84. [PMID: 37046242 PMCID: PMC10099890 DOI: 10.1186/s12933-023-01803-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/16/2023] [Indexed: 04/14/2023] Open
Abstract
AIM Obesity is linked to cardiometabolic diseases, however non-obese individuals are also at risk for type 2 diabetes (T2D) and cardiovascular disease (CVD). White adipose tissue (WAT) is known to play a role in both T2D and CVD, but the contribution of WAT inflammatory status especially in non-obese patients with cardiometabolic diseases is less understood. Therefore, we aimed to find associations between WAT inflammatory status and cardiometabolic diseases in non-obese individuals. METHODS In a population-based cohort containing non-obese healthy (n = 17), T2D (n = 16), CVD (n = 18), T2D + CVD (n = 19) individuals, seventeen different cytokines were measured in WAT and in circulation. In addition, 13-color flow cytometry profiling was employed to phenotype the immune cells. Human T cell line (Jurkat T cells) was stimulated by rCCL18, and conditioned media (CM) was added to the in vitro cultures of human adipocytes. Lipolysis was measured by glycerol release. Blocking antibodies against IFN-γ and TGF-β were used in vitro to prove a role for these cytokines in CCL18-T-cell-adipocyte lipolysis regulation axis. RESULTS In CVD, T2D and CVD + T2D groups, CCL18 and CD4+ T cells were upregulated significantly compared to healthy controls. WAT CCL18 secretion correlated with the amounts of WAT CD4+ T cells, which also highly expressed CCL18 receptors suggesting that WAT CD4+ T cells are responders to this chemokine. While direct addition of rCCL18 to mature adipocytes did not alter the adipocyte lipolysis, CM from CCL18-treated T cells increased glycerol release in in vitro cultures of adipocytes. IFN-γ and TGF-β secretion was significantly induced in CM obtained from T cells treated with CCL18. Blocking these cytokines in CM, prevented CM-induced upregulation of adipocyte lipolysis. CONCLUSION We suggest that in T2D and CVD, increased production of CCL18 recruits and activates CD4+ T cells to secrete IFN-γ and TGF-β. This, in turn, promotes adipocyte lipolysis - a possible risk factor for cardiometabolic diseases.
Collapse
Affiliation(s)
- Narmadha Subramanian
- Lipid laboratory, Unit of Endocrinology, Dept. of Medicine Huddinge, Karolinska Institutet, Stockholm, 141 86, Sweden
| | - Kaisa Hofwimmer
- Lipid laboratory, Unit of Endocrinology, Dept. of Medicine Huddinge, Karolinska Institutet, Stockholm, 141 86, Sweden
| | - Beatriz Tavira
- Lipid laboratory, Unit of Endocrinology, Dept. of Medicine Huddinge, Karolinska Institutet, Stockholm, 141 86, Sweden
| | - Lucas Massier
- Lipid laboratory, Unit of Endocrinology, Dept. of Medicine Huddinge, Karolinska Institutet, Stockholm, 141 86, Sweden
| | - Daniel P Andersson
- Lipid laboratory, Unit of Endocrinology, Dept. of Medicine Huddinge, Karolinska Institutet, Stockholm, 141 86, Sweden
| | - Peter Arner
- Lipid laboratory, Unit of Endocrinology, Dept. of Medicine Huddinge, Karolinska Institutet, Stockholm, 141 86, Sweden
| | - Jurga Laurencikiene
- Lipid laboratory, Unit of Endocrinology, Dept. of Medicine Huddinge, Karolinska Institutet, Stockholm, 141 86, Sweden.
| |
Collapse
|
4
|
An integrated single cell and spatial transcriptomic map of human white adipose tissue. Nat Commun 2023; 14:1438. [PMID: 36922516 PMCID: PMC10017705 DOI: 10.1038/s41467-023-36983-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
To date, single-cell studies of human white adipose tissue (WAT) have been based on small cohort sizes and no cellular consensus nomenclature exists. Herein, we performed a comprehensive meta-analysis of publicly available and newly generated single-cell, single-nucleus, and spatial transcriptomic results from human subcutaneous, omental, and perivascular WAT. Our high-resolution map is built on data from ten studies and allowed us to robustly identify >60 subpopulations of adipocytes, fibroblast and adipogenic progenitors, vascular, and immune cells. Using these results, we deconvolved spatial and bulk transcriptomic data from nine additional cohorts to provide spatial and clinical dimensions to the map. This identified cell-cell interactions as well as relationships between specific cell subtypes and insulin resistance, dyslipidemia, adipocyte volume, and lipolysis upon long-term weight changes. Altogether, our meta-map provides a rich resource defining the cellular and microarchitectural landscape of human WAT and describes the associations between specific cell types and metabolic states.
Collapse
|
5
|
Cruz-García EM, Frigolet ME, Canizales-Quinteros S, Gutiérrez-Aguilar R. Differential Gene Expression of Subcutaneous Adipose Tissue among Lean, Obese, and after RYGB (Different Timepoints): Systematic Review and Analysis. Nutrients 2022; 14:nu14224925. [PMID: 36432612 PMCID: PMC9693162 DOI: 10.3390/nu14224925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022] Open
Abstract
The main roles of adipose tissue include triglycerides storage and adipokine secretion, which regulate energy balance and inflammation status. In obesity, adipocyte dysfunction leads to proinflammatory cytokine production and insulin resistance. Bariatric surgery is the most effective treatment for obesity, the gold-standard technique being Roux-en-Y gastric bypass (RYGB). Since metabolic improvements after RYGB are clear, a better understanding of adipose tissue molecular modifications could be derived from this study. Thus, the aim of this systematic review was to find differentially expressed genes in subcutaneous adipose tissue of lean, obese and post-RYGB (distinct timepoints). To address this objective, publications from 2015-2022 reporting gene expression (candidate genes or transcriptomic approach) of subcutaneous adipose tissue from lean and obese individuals before and after RGYB were searched in PubMed, Elsevier, and Springer Link. Excluded publications were reviews, studies analyzing serum, other types of tissues, or bariatric procedures. A risk-of-bias summary was created for each paper using Robvis, to finally include 17 studies. Differentially expressed genes in post-RYGB vs. obese and lean vs. obese were obtained and the intersection among these groups was used for analysis and gene classification by metabolic pathway. Results showed that the lean state as well as the post-RYGB is similar in terms of increased expression of insulin-sensitizing molecules, inducing lipogenesis over lipolysis and downregulating leukocyte activation, cytokine production and other factors that promote inflammation. Thus, massive weight loss and metabolic improvements after RYGB are accompanied by gene expression modifications reverting the "adipocyte dysfunction" phenomenon observed in obesity conditions.
Collapse
Affiliation(s)
- Elena Marisol Cruz-García
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México “Federico Gómez”, Mexico City 06720, Mexico
| | - María E. Frigolet
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México “Federico Gómez”, Mexico City 06720, Mexico
| | - Samuel Canizales-Quinteros
- Unidad de Genόmica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/Instituto Nacional de Medicina Genόmica (INMEGEN), Mexico City 14610, Mexico
| | - Ruth Gutiérrez-Aguilar
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México “Federico Gómez”, Mexico City 06720, Mexico
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
- Correspondence: ; Tel.: +52-5552289917 (ext. 4509)
| |
Collapse
|
6
|
Rydén M, Andersson DP, Kotopouli MI, Stenberg E, Näslund E, Thorell A, Sørensen TIA, Arner P. Lipolysis defect in people with obesity who undergo metabolic surgery. J Intern Med 2022; 292:667-678. [PMID: 35670497 PMCID: PMC9540545 DOI: 10.1111/joim.13527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Cross-sectional studies demonstrate that catecholamine stimulation of fat cell lipolysis is blunted in obesity. We investigated whether this defect persists after substantial weight loss has been induced by metabolic surgery, and whether it is related to the outcome. DESIGN/METHODS Patients with obesity not able to successfully reduce body weight by conventional means (n = 126) were investigated before and 5 years after Roux-en-Y gastric bypass surgery (RYGB). They were compared with propensity-score matched subjects selected from a control group (n = 1017), and with the entire group after adjustment for age, sex, body mass index (BMI), fat cell volume and other clinical parameters. Catecholamine-stimulated lipolysis (glycerol release) was investigated in isolated fat cells using noradrenaline (natural hormone) or isoprenaline (synthetic beta-adrenoceptor agonist). RESULTS Following RYGB, BMI was reduced from 39.9 (37.5-43.5) (median and interquartile range) to 29.5 (26.7-31.9) kg/m2 (p < 0.0001). The post-RYGB patients had about 50% lower lipolysis rates compared with the matched and total series of controls (p < 0.0005). Nordrenaline activation of lipolysis at baseline was associated with the RYGB effect; those with high lipolysis activation (upper tertile) lost 30%-45% more in body weight, BMI or fat mass than those with low (bottom tertile) initial lipolysis activation (p < 0.0007). CONCLUSION Patients with obesity requiring metabolic surgery have impaired ability of catecholamines to stimulate lipolysis, which remains despite long-term normalization of body weight by RYGB. Furthermore, preoperative variations in the ability of catecholamines to activate lipolysis may predict the long-term reduction in body weight and fat mass.
Collapse
Affiliation(s)
- Mikael Rydén
- Department of Medicine (H7), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Daniel P Andersson
- Department of Medicine (H7), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Maria I Kotopouli
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Erik Stenberg
- Department of Surgery, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Erik Näslund
- Division of Surgery, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Anders Thorell
- Department of Clinical Sciences, Danderyds Hospital, Karolinska Institutet, Stockholm, Sweden.,Department of Surgery, Ersta Hospital, Stockholm, Sweden
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Centre for Basic Metabolic Research and Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Arner
- Department of Medicine (H7), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Arner P, Andersson DP, Arner E, Rydén M, Kerr AG. Subcutaneous adipose tissue expansion mechanisms are similar in early and late onset overweight/obesity. Int J Obes (Lond) 2022; 46:1196-1203. [PMID: 35228658 PMCID: PMC9151387 DOI: 10.1038/s41366-022-01102-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND/OBJECTIVE The development of overweight/obesity associates with alterations in white adipose tissue (WAT) cellularity (fat cell size/number) and lipid metabolism, in particular lipolysis. If these changes differ between early/juvenile (EOO < 18 years of age) or late onset overweight/obesity (LOO) is unknown and was presently examined. SUBJECTS/METHODS We included 439 subjects with validated information on body mass index (BMI) at 18 years of age. Using this information and current BMI, subjects were divided into never overweight/obese (BMI < 25 kg/m2), EOO and LOO. Adipocyte size, number, morphology (size in relation to body fat) and lipolysis were determined in subcutaneous abdominal WAT. Body composition and WAT distribution was assessed by dual-X-ray absorptiometry. RESULTS Compared with never overweight/obese, EOO and LOO displayed larger WAT amounts in all examined depots, which in subcutaneous WAT was explained by a combination of increased size and number of fat cells in EOO and LOO. EOO had 40% larger subcutaneous fat mass than LOO (p < 0.0001). Visceral WAT mass, WAT morphology and lipolysis did not differ between EOO and LOO except for minor differences in men between the two obesity groups. On average, the increase in BMI per year was 57% higher in subjects with EOO compared to LOO (p < 0.0001). CONCLUSION Early onset overweight/obesity causes a more rapid and pronounced accumulation of subcutaneous WAT than adult onset. However, fat mass expansion measures including WAT cellularity, morphology and fat cell lipolysis do not differ in an important way suggesting that similar mechanisms of WAT growth operate in EOO and LOO.
Collapse
Affiliation(s)
- Peter Arner
- Department of Medicine (H7), Karolinska Institutet at Karolinska University Hospital Huddinge, Center for Metabolism and Endocrinology, 14186, Stockholm, Sweden.
| | - Daniel P Andersson
- Department of Medicine (H7), Karolinska Institutet at Karolinska University Hospital Huddinge, Center for Metabolism and Endocrinology, 14186, Stockholm, Sweden
| | - Erik Arner
- Department of Medicine (H7), Karolinska Institutet at Karolinska University Hospital Huddinge, Center for Metabolism and Endocrinology, 14186, Stockholm, Sweden
- GSK, Gunnels Wood Rd, Stevenage, SG1 2NY, United Kingdom
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet at Karolinska University Hospital Huddinge, Center for Metabolism and Endocrinology, 14186, Stockholm, Sweden
| | - Alastair G Kerr
- Department of Medicine (H7), Karolinska Institutet at Karolinska University Hospital Huddinge, Center for Metabolism and Endocrinology, 14186, Stockholm, Sweden.
| |
Collapse
|
8
|
Kulyté A, Aman A, Strawbridge RJ, Arner P, Dahlman IA. Genome-Wide Association Study Identifies Genetic Loci Associated With Fat Cell Number and Overlap With Genetic Risk Loci for Type 2 Diabetes. Diabetes 2022; 71:1350-1362. [PMID: 35320353 PMCID: PMC9163556 DOI: 10.2337/db21-0804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 03/17/2022] [Indexed: 11/13/2022]
Abstract
Interindividual differences in generation of new fat cells determine body fat and type 2 diabetes risk. In the GENetics of Adipocyte Lipolysis (GENiAL) cohort, which consists of participants who have undergone abdominal adipose biopsy, we performed a genome-wide association study (GWAS) of fat cell number (n = 896). Candidate genes from the genetic study were knocked down by siRNA in human adipose-derived stem cells. We report 318 single nucleotide polymorphisms (SNPs) and 17 genetic loci displaying suggestive (P < 1 × 10-5) association with fat cell number. Two loci pass threshold for GWAS significance, on chromosomes 2 (lead SNP rs149660479-G) and 7 (rs147389390-deletion). We filtered for fat cell number-associated SNPs (P < 1.00 × 10-5) using evidence of genotype-specific expression. Where this was observed we selected genes for follow-up investigation and hereby identified SPATS2L and KCTD18 as regulators of cell proliferation consistent with the genetic data. Furthermore, 30 reported type 2 diabetes-associated SNPs displayed nominal and consistent associations with fat cell number. In functional follow-up of candidate genes, RPL8, HSD17B12, and PEPD were identified as displaying effects on cell proliferation consistent with genetic association and gene expression findings. In conclusion, findings presented herein identify SPATS2L, KCTD18, RPL8, HSD17B12, and PEPD of potential importance in controlling fat cell numbers (plasticity), the size of body fat, and diabetes risk.
Collapse
Affiliation(s)
- Agné Kulyté
- Lipid Laboratory, Endocrinology Unit, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Alisha Aman
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Rona J. Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, U.K
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Peter Arner
- Lipid Laboratory, Endocrinology Unit, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Ingrid A. Dahlman
- Lipid Laboratory, Endocrinology Unit, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
- Corresponding author: Ingrid A. Dahlman,
| |
Collapse
|
9
|
Kerr AG, Wang Z, Wang N, Kwok KHM, Jalkanen J, Ludzki A, Lecoutre S, Langin D, Bergo MO, Dahlman I, Mim C, Arner P, Gao H. The long noncoding RNA ADIPINT regulates human adipocyte metabolism via pyruvate carboxylase. Nat Commun 2022; 13:2958. [PMID: 35618718 PMCID: PMC9135762 DOI: 10.1038/s41467-022-30620-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 05/04/2022] [Indexed: 12/27/2022] Open
Abstract
The pleiotropic function of long noncoding RNAs is well recognized, but their direct role in governing metabolic homeostasis is less understood. Here, we describe a human adipocyte-specific lncRNA, ADIPINT, that regulates pyruvate carboxylase, a pivotal enzyme in energy metabolism. We developed an approach, Targeted RNA-protein identification using Orthogonal Organic Phase Separation, which identifies that ADIPINT binds to pyruvate carboxylase and validated the interaction with electron microscopy. ADIPINT knockdown alters the interactome and decreases the abundance and enzymatic activity of pyruvate carboxylase in the mitochondria. Reduced ADIPINT or pyruvate carboxylase expression lowers adipocyte lipid synthesis, breakdown, and lipid content. In human white adipose tissue, ADIPINT expression is increased in obesity and linked to fat cell size, adipose insulin resistance, and pyruvate carboxylase activity. Thus, we identify ADIPINT as a regulator of lipid metabolism in human white adipocytes, which at least in part is mediated through its interaction with pyruvate carboxylase. Adipocyte-expressed long non-coding RNAs (lncRNAs) have been shown to regulate the transcription of genes involved in lipid metabolism. Here the authors describe a human adipocyte-specific lncRNA, ADIPINT, which regulates lipid metabolism in white adipocytes in part through its interaction with the metabolic enzyme pyruvate carboxylase.
Collapse
Affiliation(s)
- Alastair G Kerr
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, 141 86, Sweden
| | - Zuoneng Wang
- Department of Biomedical Engineering and Health Systems, Royal Technical Institute, Stockholm, Sweden
| | - Na Wang
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, 141 86, Sweden
| | - Kelvin H M Kwok
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, 141 86, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 141 83, Sweden
| | - Jutta Jalkanen
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, 141 86, Sweden
| | - Alison Ludzki
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, 141 86, Sweden
| | - Simon Lecoutre
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, 141 86, Sweden
| | - Dominique Langin
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (Inserm), Université de Toulouse, UPS, UMR1297, Toulouse, France.,Department of Biochemistry, Toulouse University Hospitals, CHU Toulouse, Toulouse, France
| | - Martin O Bergo
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 141 83, Sweden
| | - Ingrid Dahlman
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, 141 86, Sweden
| | - Carsten Mim
- Department of Biomedical Engineering and Health Systems, Royal Technical Institute, Stockholm, Sweden
| | - Peter Arner
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, 141 86, Sweden.
| | - Hui Gao
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 141 83, Sweden.
| |
Collapse
|
10
|
Influence of NAFLD and bariatric surgery on hepatic and adipose tissue mitochondrial biogenesis and respiration. Nat Commun 2022; 13:2931. [PMID: 35614135 PMCID: PMC9132900 DOI: 10.1038/s41467-022-30629-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
Impaired mitochondrial oxidative phosphorylation (OXPHOS) in liver tissue has been hypothesised to contribute to the development of nonalcoholic steatohepatitis in patients with nonalcoholic fatty liver disease (NAFLD). It is unknown whether OXPHOS capacities in human visceral (VAT) and subcutaneous adipose tissue (SAT) associate with NAFLD severity and how hepatic OXPHOS responds to improvement in NAFLD. In biopsies sampled from 62 patients with obesity undergoing bariatric surgery and nine control subjects without obesity we demonstrate that OXPHOS is reduced in VAT and SAT while increased in the liver in patients with obesity when compared with control subjects without obesity, but this was independent of NAFLD severity. In repeat liver biopsy sampling in 21 patients with obesity 12 months after bariatric surgery we found increased hepatic OXPHOS capacity and mitochondrial DNA/nuclear DNA content compared with baseline. In this work we show that obesity has an opposing association with mitochondrial respiration in adipose- and liver tissue with no overall association with NAFLD severity, however, bariatric surgery increases hepatic OXPHOS and mitochondrial biogenesis. Impaired mitochondrial function in liver tissue may contribute to the pathogenesis and disease progression of nonalcoholic fatty liver disease (NAFLD). Here the authors report that patients with obesity have lower mitochondrial capacity in adipose tissues but higher capacity in the liver, without overall associations to NAFLD severity, and that bariatric surgery increases hepatic mitochondrial respiration and mitochondrial biogenesis.
Collapse
|
11
|
Gasmi A, Bjørklund G, Mujawdiya PK, Semenova Y, Dosa A, Piscopo S, Pen JJ, Gasmi Benahmed A, Costea DO. Gut microbiota in bariatric surgery. Crit Rev Food Sci Nutr 2022; 63:9299-9314. [PMID: 35531940 DOI: 10.1080/10408398.2022.2067116] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Gut microbes share a symbiotic relationship with humans and perform several metabolic and physiological functions essential for human survival. It has been established in several scientific studies that obesity and other metabolic complications are always associated with disturbed gut microbiota profile, also called gut dysbiosis. In recent years, bariatric surgery has become a treatment of choice for weight loss, and it forms an important part of obesity management strategies across the globe. Interestingly, bariatric surgery has been shown to alter gut microbiota profile and synthesize short-chain fatty acids by gut microbes. In other words, gut microbes play a crucial role in better clinical outcomes associated with bariatric surgery. In addition, gut microbes are important in reducing weight and lowering the adverse events post-bariatric surgery. Therefore, several prebiotics, probiotics and postbiotics are recommended for patients who underwent bariatric surgery procedures for better clinical outcomes. The present review aims to understand the possible association between gut microbes and bariatric surgery and present scientific evidence showing the beneficial role of gut microbes in improving therapeutic outcomes of bariatric surgery.
Collapse
Affiliation(s)
- Amin Gasmi
- Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, Villeurbanne, France
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Claude Bernard, Villeurbanne, France
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| | | | - Yuliya Semenova
- Department of Neurology, Ophthalmology, and ENT, Semey Medical University, Semey, Kazakhstan
- CONEM Kazakhstan Environmental Health and Safety Research Group, Semey Medical University, Semey, Kazakhstan
| | | | - Salva Piscopo
- Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, Villeurbanne, France
- Department of Nutritional Research and Development, Nutri-Logics SA, Weiswampach, Luxembourg
| | - Joeri J Pen
- Diabetes Clinic, Department of Internal Medicine, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Nutrition, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Asma Gasmi Benahmed
- Académie Internationale de Médecine Dentaire Intégrative, Paris, France
- Université Claude Bernard -Lyon 1, Villeurbanne, France
| | | |
Collapse
|
12
|
Abstract
Recent technological developments have allowed determination of the age of fat cells and their lipids in adult humans. In contrast to prior views, this has demonstrated a high turnover rate of the fat cells (10%/year) and their unilocular lipid droplets (six times/10 years). Fat cell turnover is increased in obesity and when adipose tissue is composed of many but small adipocytes (hyperplasia, a benign adipose phenotype). While fat mass gain increases adipocyte number and size, only the latter is altered (decreased) after weight loss, which may facilitate weight regain. Fat cell lipid turnover is attenuated in subjects with excess body fat. In the subcutaneous region, this dysregulation occurs already in the overweight state while in the visceral depot, it is only observed in severe obesity. This may explain why the latter depot is particularly pernicious in the overweight/obese state as it allows for more rapid lipid fluxes between visceral fat and the liver. Adipose lipid turnover decreases during ageing due to impaired breakdown (lipolysis) of stored triglycerides. If this decline is not compensated by reduced adipocyte lipid uptake, bodyweight will increase over time. In concordance with this, low lipolysis rates are a risk factor for future weight gain and glucose intolerance. Adipose lipid turnover is also decreased in insulin resistance and certain forms of dyslipidemia. Altogether, adult human adipose tissue is in a highly dynamic state. Alterations in the turnover of fat cells and their lipids are therefore novel factors to consider in the pathophysiology of common metabolic disorders.
Collapse
Affiliation(s)
- Peter Arner
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Rydén
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
13
|
Rodríguez‐Barrueco R, Latorre J, Devis‐Jáuregui L, Lluch A, Bonifaci N, Llobet FJ, Olivan M, Coll‐Iglesias L, Gassner K, Davis ML, Moreno‐Navarrete JM, Castells‐Nobau A, Plata‐Peña L, Dalmau‐Pastor M, Höring M, Liebisch G, Olkkonen VM, Arnoriaga‐Rodríguez M, Ricart W, Fernández‐Real JM, Silva JM, Ortega FJ, Llobet‐Navas D. A microRNA Cluster Controls Fat Cell Differentiation and Adipose Tissue Expansion By Regulating SNCG. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104759. [PMID: 34898027 PMCID: PMC8811811 DOI: 10.1002/advs.202104759] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Indexed: 05/08/2023]
Abstract
The H19X-encoded miR-424(322)/503 cluster regulates multiple cellular functions. Here, it is reported for the first time that it is also a critical linchpin of fat mass expansion. Deletion of this miRNA cluster in mice results in obesity, while increasing the pool of early adipocyte progenitors and hypertrophied adipocytes. Complementary loss and gain of function experiments and RNA sequencing demonstrate that miR-424(322)/503 regulates a conserved genetic program involved in the differentiation and commitment of white adipocytes. Mechanistically, it is demonstrated that miR-424(322)/503 targets γ-Synuclein (SNCG), a factor that mediates this program rearrangement by controlling metabolic functions in fat cells, allowing adipocyte differentiation and adipose tissue enlargement. Accordingly, diminished miR-424(322) in mice and obese humans co-segregate with increased SNCG in fat and peripheral blood as mutually exclusive features of obesity, being normalized upon weight loss. The data unveil a previously unknown regulatory mechanism of fat mass expansion tightly controlled by the miR-424(322)/503 through SNCG.
Collapse
Affiliation(s)
- Ruth Rodríguez‐Barrueco
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Anatomy UnitDepartment of Pathology and Experimental TherapySchool of MedicineUniversity of Barcelona (UB)L'Hospitalet de Llobregat08907Spain
| | - Jessica Latorre
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Laura Devis‐Jáuregui
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
| | - Aina Lluch
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
| | - Nuria Bonifaci
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos III, (ISCIII)Madrid28029Spain
| | - Francisco J. Llobet
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
| | - Mireia Olivan
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Anatomy UnitDepartment of Pathology and Experimental TherapySchool of MedicineUniversity of Barcelona (UB)L'Hospitalet de Llobregat08907Spain
| | - Laura Coll‐Iglesias
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
| | - Katja Gassner
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos III, (ISCIII)Madrid28029Spain
| | - Meredith L. Davis
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Department of PathologyDuke University School of MedicineDurhamNC27710USA
| | - José M. Moreno‐Navarrete
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Anna Castells‐Nobau
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
| | - Laura Plata‐Peña
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
| | - Miki Dalmau‐Pastor
- Anatomy UnitDepartment of Pathology and Experimental TherapySchool of MedicineUniversity of Barcelona (UB)L'Hospitalet de Llobregat08907Spain
- MIFAS by GRECMIP (Minimally Invasive Foot and Ankle Society)Merignac33700France
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory MedicineRegensburg University HospitalRegensburg93053Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory MedicineRegensburg University HospitalRegensburg93053Germany
| | - Vesa M. Olkkonen
- Minerva Foundation Institute for Medical Research (Biomedicum 2U)and Department of AnatomyFaculty of MedicineUniversity of HelsinkiHelsinki00290Finland
| | - Maria Arnoriaga‐Rodríguez
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Wifredo Ricart
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - José M. Fernández‐Real
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - José M. Silva
- Department of PathologyIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| | - Francisco J. Ortega
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - David Llobet‐Navas
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos III, (ISCIII)Madrid28029Spain
| |
Collapse
|
14
|
LONG-TERM IMPROVEMENT OF ADIPOCYTE INSULIN ACTION DURING BODY WEIGHT RELAPSE AFTER BARIATRIC SURGERY: A LONGITUDINAL COHORT STUDY. Surg Obes Relat Dis 2022; 18:683-692. [DOI: 10.1016/j.soard.2022.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 12/19/2022]
|
15
|
Ye RZ, Richard G, Gévry N, Tchernof A, Carpentier AC. Fat Cell Size: Measurement Methods, Pathophysiological Origins, and Relationships With Metabolic Dysregulations. Endocr Rev 2022; 43:35-60. [PMID: 34100954 PMCID: PMC8755996 DOI: 10.1210/endrev/bnab018] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Indexed: 11/19/2022]
Abstract
The obesity pandemic increasingly causes morbidity and mortality from type 2 diabetes, cardiovascular diseases and many other chronic diseases. Fat cell size (FCS) predicts numerous obesity-related complications such as lipid dysmetabolism, ectopic fat accumulation, insulin resistance, and cardiovascular disorders. Nevertheless, the scarcity of systematic literature reviews on this subject is compounded by the use of different methods by which FCS measurements are determined and reported. In this paper, we provide a systematic review of the current literature on the relationship between adipocyte hypertrophy and obesity-related glucose and lipid dysmetabolism, ectopic fat accumulation, and cardiovascular disorders. We also review the numerous mechanistic origins of adipocyte hypertrophy and its relationship with metabolic dysregulation, including changes in adipogenesis, cell senescence, collagen deposition, systemic inflammation, adipokine secretion, and energy balance. To quantify the effect of different FCS measurement methods, we performed statistical analyses across published data while controlling for body mass index, age, and sex.
Collapse
Affiliation(s)
- Run Zhou Ye
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Gabriel Richard
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Nicolas Gévry
- Department of Biology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - André Tchernof
- Québec Heart and Lung Research Institute, Laval University, Québec, Québec, Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
16
|
Santini S, Vionnet N, Pasquier J, Suter M, Hans D, Gonzalez-Rodriguez E, Pitteloud N, Favre L. Long-term body composition improvement in post-menopausal women following bariatric surgery: a cross-sectional and case-control study. Eur J Endocrinol 2022; 186:255-263. [PMID: 34879003 PMCID: PMC8789027 DOI: 10.1530/eje-21-0895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/08/2021] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Bariatric surgery (BS) induces loss of body fat mass (FM) with an inexorable loss of lean mass (LM). Menopause leads to deleterious changes in body composition (BC) related to estrogen deficiency including LM loss and increase in total and visceral adipose tissue (VAT). This study aims to describe the long-term weight evolution of post-menopausal women after Roux-en-Y gastric bypass (RYGB) and to compare the BC between BS patients vs post-menopausal non-operated women. DESIGN Cross-sectional study of 60 post-menopausal women who underwent RYGB ≥2 years prior to the study with nested case-control design. METHODS Post-menopausal BS women were matched for age and BMI with controls. Both groups underwent DXA scan, lipids and glucose metabolism markers assessment. RESULTS Median follow-up was 7.5 (2-18) years. Percentage of total weight loss (TWL%) was 28.5 ± 10%. After RYGB, LM percentage of body weight (LM%) was positively associated with TWL% and negatively associated with nadir weight. Forty-one post-BS women were age- and BMI-matched with controls. Post-BS patients showed higher LM% (57.7% (±8%) vs 52.5% (±5%), P = 0.001), reduced FM% (39.4% (±8.4%) vs 45.9% (±5.4%), P < 0.01) and lower VAT (750.6 g (±496) vs 1295.3 g (±688), P < 0.01) with no difference in absolute LM compared to controls. While post-BS women showed a better lipid profile compared to controls, no difference was found in glucose markers. CONCLUSIONS Post-menopausal women after RYGB have a lower FM and VAT, preserved LM and a better lipid profile compared to controls. Weight loss after RYGB seems to have a persistent positive impact on metabolic health.
Collapse
Affiliation(s)
- Sara Santini
- Division of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Nathalie Vionnet
- Division of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Jérôme Pasquier
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Michel Suter
- Department of Visceral Surgery, Lausanne University Hospital, Lausanne, Switzerland
- Department of Surgery, Riviera-Chablais Hospital, Rennaz, Switzerland
- Faculty of Biology and medicine, University of Lausanne, Lausanne, Switzerland
| | - Didier Hans
- Faculty of Biology and medicine, University of Lausanne, Lausanne, Switzerland
- Interdisciplinary Center for Bone Diseases, Lausanne University Hospital, Lausanne, Switzerland
| | - Elena Gonzalez-Rodriguez
- Faculty of Biology and medicine, University of Lausanne, Lausanne, Switzerland
- Interdisciplinary Center for Bone Diseases, Lausanne University Hospital, Lausanne, Switzerland
| | - Nelly Pitteloud
- Division of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Faculty of Biology and medicine, University of Lausanne, Lausanne, Switzerland
| | - Lucie Favre
- Division of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Faculty of Biology and medicine, University of Lausanne, Lausanne, Switzerland
- Correspondence should be addressed to L Favre;
| |
Collapse
|
17
|
Zhu L, Liu L. New Insights Into the Interplay Among Autophagy, the NLRP3 Inflammasome and Inflammation in Adipose Tissue. Front Endocrinol (Lausanne) 2022; 13:739882. [PMID: 35432210 PMCID: PMC9008752 DOI: 10.3389/fendo.2022.739882] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity is a feature of metabolic syndrome with chronic inflammation in obese subjects, characterized by adipose tissue (AT) expansion, proinflammatory factor overexpression, and macrophage infiltration. Autophagy modulates inflammation in the enlargement of AT as an essential step for maintaining the balance in energy metabolism and waste elimination. Signaling originating from dysfunctional AT, such as AT containing hypertrophic adipocytes and surrounding macrophages, activates NOD-like receptor family 3 (NLRP3) inflammasome. There are interactions about altered autophagy and NLRP3 inflammasome activation during the progress in obesity. We summarize the current studies and potential mechanisms associated with autophagy and NLRP3 inflammasome in AT inflammation and aim to provide further evidence for research on obesity and obesity-related complications.
Collapse
Affiliation(s)
- Liyuan Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Ling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
- *Correspondence: Ling Liu,
| |
Collapse
|
18
|
Impact of Bariatric Surgery on Adipose Tissue Biology. J Clin Med 2021; 10:jcm10235516. [PMID: 34884217 PMCID: PMC8658722 DOI: 10.3390/jcm10235516] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023] Open
Abstract
Bariatric surgery (BS) procedures are actually the most effective intervention to help subjects with severe obesity achieve significant and sustained weight loss. White adipose tissue (WAT) is increasingly recognized as the largest endocrine organ. Unhealthy WAT expansion through adipocyte hypertrophy has pleiotropic effects on adipocyte function and promotes obesity-associated metabolic complications. WAT dysfunction in obesity encompasses an altered adipokine secretome, unresolved inflammation, dysregulated autophagy, inappropriate extracellular matrix remodeling and insufficient angiogenic potential. In the last 10 years, accumulating evidence suggests that BS can improve the WAT function beyond reducing the fat depot sizes. The causal relationships between improved WAT function and the health benefits of BS merits further investigation. This review summarizes the current knowledge on the short-, medium- and long-term outcomes of BS on the WAT composition and function.
Collapse
|
19
|
Hjortebjerg R, Bojsen-Møller KN, Søeby M, Oxvig C, Madsbad S, Frystyk J. Metabolic improvement after gastric bypass correlates with changes in IGF-regulatory proteins stanniocalcin-2 and IGFBP-4. Metabolism 2021; 124:154886. [PMID: 34506805 DOI: 10.1016/j.metabol.2021.154886] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/13/2021] [Accepted: 09/01/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Pregnancy-associated plasma protein-A (PAPP-A) is an enzyme that increases IGF-activity through cleavage of IGF-binding proteins (IGFBPs), primarily IGFBP-4, whereby bound IGF-I becomes released as a free molecule. The enzymatic activity of PAPP-A is irreversibly suppressed by the glycoprotein stanniocalcin-2 (STC2). Pre-clinical and clinical studies suggest that the STC2 - PAPP-A - IGFBP-4 axis is important in controlling local IGF-action. STC2, PAPP-A and IGFBP-4 are expressed in adipose tissue, and as bariatric surgery markedly reduces the amount of fat, we found it relevant to study the impact of Roux-en-Y gastric bypass (RYGB) on circulating concentrations of this IGF-regulatory network. METHODS Analysis of fasting blood samples from 20 obese subjects, hereof 10 with preoperative type 2 diabetes, investigated before RYGB, and 1 week, 3 months and 12 months post-surgery. Members of the IGF-system were analyzed by immunoassays, bioactive IGF by cell-based IGF-I receptor activation assay. We compared changes in IGF-system components with changes in fasting plasma insulin and glucose, and HbA1c. RESULTS PAPP-A remained unchanged, but STC2 decreased following RYGB (p < 0.05). The PAPP-A substrate IGFBP-4 declined (p < 0.01), whereas levels of PAPP-A specific IGFBP-4 fragments increased (p < 0.05), indicating an increased PAPP-A enzymatic activity post-RYGB. Further, the reduction in intact IGFBP-4 correlated with increased levels of bioactive IGF (p < 0.05). In multivariable regression analyses, an improved glucose metabolism correlated with reductions in STC2 and IGFBP-4, and with increases in bioactive IGF and IGF-I (p < 0.05). CONCLUSION After 12 months, RYGB caused reduced serum concentrations of intact IGFBP-4 and STC2, whereas serum PAPP-A remained at pre-operative levels. However, concentrations of PAPP-A generated IGFBP-4 fragments increased, pointing to an overall increased PAPP-A enzymatic activity following RYGB. Notably, reductions in intact IGFBP-4 and STC2 associated with improvements in glucose metabolism. Therefore, we propose that STC2 and IGFBP-4 are involved in the metabolic improvement that follows RYGB.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital & Department of Clinical Research, Faculty of Health, University of Southern Denmark, Odense, Denmark; Steno Diabetes Center Odense, Odense University Hospital & Department of Clinical Research, Faculty of Health, University of Southern Denmark, Denmark; Medical Research Laboratory, Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | | | - Mette Søeby
- Medical Research Laboratory, Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre Hospital, Hvidovre, Denmark
| | - Jan Frystyk
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital & Department of Clinical Research, Faculty of Health, University of Southern Denmark, Odense, Denmark; Medical Research Laboratory, Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
20
|
Mileti E, Kwok KHM, Andersson DP, Mathelier A, Raman A, Bäckdahl J, Jalkanen J, Massier L, Thorell A, Gao H, Arner P, Mejhert N, Daub CO, Rydén M. Human White Adipose Tissue Displays Selective Insulin Resistance in the Obese State. Diabetes 2021; 70:1486-1497. [PMID: 33863803 DOI: 10.2337/db21-0001] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 04/14/2021] [Indexed: 11/13/2022]
Abstract
Selective hepatic insulin resistance is a feature of obesity and type 2 diabetes. Whether similar mechanisms operate in white adipose tissue (WAT) of those with obesity and to what extent these are normalized by weight loss are unknown. We determined insulin sensitivity by hyperinsulinemic euglycemic clamp and insulin response in subcutaneous WAT by RNA sequencing in 23 women with obesity before and 2 years after bariatric surgery. To control for effects of surgery, women postsurgery were matched to never-obese women. Multidimensional analyses of 138 samples allowed us to classify the effects of insulin into three distinct expression responses: a common set was present in all three groups and included genes encoding several lipid/cholesterol biosynthesis enzymes; a set of obesity-attenuated genes linked to tissue remodeling and protein translation was selectively regulated in the two nonobese states; and several postobesity-enriched genes encoding proteins involved in, for example, one-carbon metabolism were only responsive to insulin in the women who had lost weight. Altogether, human WAT displays a selective insulin response in the obese state, where most genes are normalized by weight loss. This comprehensive atlas provides insights into the transcriptional effects of insulin in WAT and may identify targets to improve insulin action.
Collapse
Affiliation(s)
- Enrichetta Mileti
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Kelvin H M Kwok
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Daniel P Andersson
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anthony Mathelier
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Jesper Bäckdahl
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jutta Jalkanen
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lucas Massier
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Thorell
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Department of Surgery, Ersta Hospital, Stockholm, Sweden
| | - Hui Gao
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Peter Arner
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Niklas Mejhert
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Carsten O Daub
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Stockholm, Sweden
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
21
|
van der Kolk BW, Muniandy M, Kaminska D, Alvarez M, Ko A, Miao Z, Valsesia A, Langin D, Vaittinen M, Pääkkönen M, Jokinen R, Kaye S, Heinonen S, Virtanen KA, Andersson DP, Männistö V, Saris WH, Astrup A, Rydén M, Blaak EE, Pajukanta P, Pihlajamäki J, Pietiläinen KH. Differential Mitochondrial Gene Expression in Adipose Tissue Following Weight Loss Induced by Diet or Bariatric Surgery. J Clin Endocrinol Metab 2021; 106:1312-1324. [PMID: 33560372 PMCID: PMC8063261 DOI: 10.1210/clinem/dgab072] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 12/13/2022]
Abstract
CONTEXT Mitochondria are essential for cellular energy homeostasis, yet their role in subcutaneous adipose tissue (SAT) during different types of weight-loss interventions remains unknown. OBJECTIVE To investigate how SAT mitochondria change following diet-induced and bariatric surgery-induced weight-loss interventions in 4 independent weight-loss studies. METHODS The DiOGenes study is a European multicenter dietary intervention with an 8-week low caloric diet (LCD; 800 kcal/d; n = 261) and 6-month weight-maintenance (n = 121) period. The Kuopio Obesity Surgery study (KOBS) is a Roux-en-Y gastric bypass (RYGB) surgery study (n = 172) with a 1-year follow-up. We associated weight-loss percentage with global and 2210 mitochondria-related RNA transcripts in linear regression analysis adjusted for age and sex. We repeated these analyses in 2 studies. The Finnish CRYO study has a 6-week LCD (800-1000 kcal/d; n = 19) and a 10.5-month follow-up. The Swedish DEOSH study is a RYGB surgery study with a 2-year (n = 49) and 5-year (n = 37) follow-up. RESULTS Diet-induced weight loss led to a significant transcriptional downregulation of oxidative phosphorylation (DiOGenes; ingenuity pathway analysis [IPA] z-scores: -8.7 following LCD, -4.4 following weight maintenance; CRYO: IPA z-score: -5.6, all P < 0.001), while upregulation followed surgery-induced weight loss (KOBS: IPA z-score: 1.8, P < 0.001; in DEOSH: IPA z-scores: 4.0 following 2 years, 0.0 following 5 years). We confirmed an upregulated oxidative phosphorylation at the proteomics level following surgery (IPA z-score: 3.2, P < 0.001). CONCLUSIONS Differentially regulated SAT mitochondria-related gene expressions suggest qualitative alterations between weight-loss interventions, providing insights into the potential molecular mechanistic targets for weight-loss success.
Collapse
Affiliation(s)
- Birgitta W van der Kolk
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
| | - Maheswary Muniandy
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
| | - Dorota Kaminska
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Arthur Ko
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Zong Miao
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Armand Valsesia
- Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Dominique Langin
- Institut National de la Santé et de la Recherche Médicale (Inserm), Université Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Department of Biochemistry, Toulouse University Hospitals, France
| | - Maija Vaittinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Mirva Pääkkönen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Finland
| | - Riikka Jokinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
| | - Sanna Kaye
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
| | - Kirsi A Virtanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
- Turku PET Center, Turku University Hospital, Turku, Finland
| | - Daniel P Andersson
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Wim H Saris
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, MD Maastricht, The Netherlands
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, MD Maastricht, The Netherlands
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
- Obesity Center, Abdominal center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
The 2020 ESPEN Arvid Wretlind lecture: Metabolic response in bariatric surgery - Mechanisms and clinical implications. Clin Nutr 2021; 40:2602-2608. [PMID: 33933726 DOI: 10.1016/j.clnu.2021.03.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/09/2021] [Accepted: 03/31/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND & AIMS Owing to the "obesity-pandemic", an increasing number of individuals are in need of treatment for obesity and obesity-related disorders. For patients with severe disease, results with conventional treatment modalities such as diet regimens, physical activity, and pharmacologic agents most often lack satisfactory efficacy and/or sustainability. In contrast, bariatric surgery has been demonstrated to be associated with marked, long-term weight loss as well as resolution or improvement of co-morbid disease, in particular metabolic aberrations such as insulin resistance and type 2 diabetes. The underlying mechanisms for the effects of surgery-induced weight loss on such morbidity are incompletely understood. METHODS This article gives an updated overview of some aspects on the mechanisms involved in the improvement in metabolism in obese individuals submitted to surgery-induced weight loss. Patients undergoing Roux en-Y Gastric Bypass (RYGB) were studied before and at various times after the operation. Weight, body composition with determination of distribution of adipose tissue (DEXA), and insulin sensitivity (hyperinsulinemic clamp) was determined. In vitro; lipolytic activity and adipose morphology (fat cell size) was assessed. RESULTS Low calorie intake, rerouting of nutrients as well as loss of fat mass are all associated with improved insulin sensitivity after RYGB. In obese individuals, an increase in lipolytic activity in visceral adipose tissue might contribute to the association with cardiometabolic disease. However, selective reduction (omentectomy) seems not to improve insulin sensitivity or cardiometabolic risk. Adipose hyperplasia (many small cells) might be protective against metabolic abnormalities compared to hypertrophy (large cells). Preoperative fat cell size is related to improvement in insulin sensitivity after RYGB. Two years after weight loss, a change in adipose morphology to a more metabolically benign phenotype (remodeling) is seen, with a reduction of fat cell size which is correlated to the improvement in insulin sensitivity. Patients with weight regain 5 years after RYGB, still display a more benign metabolic profile compared to weight-matched controls. CONCLUSIONS Several factors contribute to the improvements in insulin sensitivity and cardiometabolic disease after surgery-induced weight loss, including low calorie intake rerouting of nutrients and loss of adipose tissue mass. Increased lipolytic activity in visceral adipose tissue as well as adipose hypertrophy relates to increased metabolic risk. RYGB-induced weight loss is associated with redistribution of adipose tissue as well as remodeling of fat cells to a more benign profile. Reduction of fat cell size might be a possible target to improve insulin sensitivity in patients with obesity in the future.
Collapse
|
23
|
Ferraz-Bannitz R, Welendorf CR, Coelho PO, Salgado W, Nonino CB, Beraldo RA, Foss-Freitas MC. Bariatric surgery can acutely modulate ER-stress and inflammation on subcutaneous adipose tissue in non-diabetic patients with obesity. Diabetol Metab Syndr 2021; 13:19. [PMID: 33593418 PMCID: PMC7887793 DOI: 10.1186/s13098-021-00623-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/02/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Bariatric surgery, especially Roux-en-Y gastric bypass (RYGB), is the most effective and durable treatment option for severe obesity. The mechanisms involving adipose tissue may be important to explain the effects of surgery. METHODS We aimed to identify the genetic signatures of adipose tissue in patients undergoing RYGB. We evaluated 13 obese, non-diabetic patients (mean age 37 years, 100% women, Body mass index (BMI) 42.2 kg/m2) one day before surgery, 3 and 6 months (M) after RYGB. RESULTS Analysis of gene expression in adipose tissue collected at surgery compared with samples collected at 3 M and 6 M Post-RYGB showed that interleukins [Interleukin 6, Tumor necrosis factor-α (TNF-α), and Monocyte chemoattractant protein-1(MCP1)] and endoplasmic reticulum stress (ERS) genes [Eukaryotic translation initiation factor 2 alpha kinase 3 (EIF2AK3) and Calreticulin (CALR)] decreased during the follow-up (P ≤ 0.01 for all). Otherwise, genes involved in energy homeostasis [Adiponectin and AMP-activated protein kinase (AMPK)], cellular response to oxidative stress [Sirtuin 1, Sirtuin 3, and Nuclear factor erythroid 2-related factor 2 (NRF2)], mitochondrial biogenesis [Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α)] and amino acids metabolism [General control nonderepressible 2 (GCN2)] increased from baseline to all other time points evaluated (P ≤ 0.01 for all). Also, expression of Peroxisome proliferator-activated receptor gamma (PPARϒ) (adipogenesis regulation) was significantly decreased after RYGB (P < 0.05). Additionally, we observed that PGC1α, SIRT1 and AMPK strongly correlated to BMI at 3 M (P ≤ 0.01 for all), as well as ADIPOQ and SIRT1 to BMI at 6 M (P ≤ 0.01 for all). CONCLUSIONS Our findings demonstrate that weight loss is associated with amelioration of inflammation and ERS and increased protection against oxidative stress in adipose tissue. These observations are strongly correlated with a decrease in BMI and essential genes that control cellular energy homeostasis, suggesting an adaptive process on a gene expression level during the caloric restriction and weight loss period after RYGB. Trial registration CAAE: 73,585,317.0.0000.5440.
Collapse
Affiliation(s)
- Rafael Ferraz-Bannitz
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil.
| | - Caroline Rossi Welendorf
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil
| | - Priscila Oliveira Coelho
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil
| | - Wilson Salgado
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto, SP, Brazil
| | - Carla Barbosa Nonino
- Laboratory of Nutrigenomic Studies, Ribeirão Preto Medical School, University of Sao Paulo (USP), Ribeirao Preto, SP, Brazil
| | - Rebeca A Beraldo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil
| | - Maria Cristina Foss-Freitas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil.
| |
Collapse
|
24
|
Rosendo-Silva D, Matafome P. Gut-adipose tissue crosstalk: A bridge to novel therapeutic targets in metabolic syndrome? Obes Rev 2021; 22:e13130. [PMID: 32815267 DOI: 10.1111/obr.13130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022]
Abstract
The gut is one of the main endocrine organs in our body, producing hormones acknowledged to play determinant roles in controlling appetite, energy balance and glucose homeostasis. One of the targets of such hormones is the adipose tissue, a major energetic reservoir, which governs overall metabolism through the secretion of adipokines. Disturbances either in nutrient and metabolic sensing and consequent miscommunication between these organs constitute a key driver to the metabolic complications clustered in metabolic syndrome. Thus, it is essential to understand how the disruption of this crosstalk might trigger adipose tissue dysfunction, a strong characteristic of obesity and insulin resistance. The beneficial effects of metabolic surgery in the amelioration of glucose homeostasis and body weight reduction allowed to understand the potential of gut signals modulation as a treatment for metabolic syndrome-related obesity and type 2 diabetes. In this review, we cover the effects of gut hormones in the modulation of adipose tissue metabolic and endocrine functions, as well as their impact in tissue plasticity. Furthermore, we discuss how the modulation of gut secretome, either through surgical procedures or pharmacological approaches, might improve adipose tissue function in obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Daniela Rosendo-Silva
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Paulo Matafome
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.,Department of Complementary Sciences, Instituto Politécnico de Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| |
Collapse
|
25
|
Vertical sleeve gastrectomy induces distinctive transcriptomic responses in liver, fat and muscle. Sci Rep 2021; 11:2310. [PMID: 33504853 PMCID: PMC7840766 DOI: 10.1038/s41598-021-81866-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Abstract
Vertical sleeve gastrectomy (VSG) is the most commonly performed bariatric/metabolic surgery, exhibiting a high rate of diabetes remission in humans. To elucidate the molecular mechanisms of VSG, we performed transcriptomic analysis of the liver, fat, and muscle in VSG mice. C57BL/6 mice fed a high-fat diet were randomly assigned to sham or VSG surgery. The sham-operated mice were fed ad libitum (sham group) or pair-fed (sham-PF group) matching their food intake to the VSG-operated mice. Comparative transcriptomic analysis of the liver, fat, and muscle using RNA sequencing was performed. VSG reduced body weight and improved glucose tolerance compared to the sham group, but not more than the sham-PF group. Improvement in fatty liver and adipose tissue inflammation was comparable between VSG and sham-PF. However, global gene expression profiles showed distinctive changes in the liver, fat, and muscle of the VSG group compared to both the sham or sham-PF groups. The liver showed the most prominent gene expression changes. Immune response-related pathways were commonly upregulated in the three organs of the VSG group compared to the sham or sham-PF. VSG induces organ-specific gene expression changes in the liver, fat, and muscle, which may play critical roles in metabolic improvements after VSG.
Collapse
|
26
|
A distribution-centered approach for analyzing human adipocyte size estimates and their association with obesity-related traits and mitochondrial function. Int J Obes (Lond) 2021; 45:2108-2117. [PMID: 34172828 PMCID: PMC8380540 DOI: 10.1038/s41366-021-00883-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/10/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Cell diameter, area, and volume are established quantitative measures of adipocyte size. However, these different adipocyte sizing parameters have not yet been directly compared regarding their distributions. Therefore, the study aimed to investigate how these adipocyte size measures differ in their distribution and assessed their correlation with anthropometry and laboratory chemistry. In addition, we were interested to investigate the relationship between fat cell size and adipocyte mitochondrial respiratory chain capacity. METHODS Subcutaneous and visceral histology-based adipocyte size estimates from 188 individuals were analyzed by applying a panel of parameters to describe the underlying cell population. Histology-based adipocyte diameter distributions were compared with adipocyte diameter distributions from collagenase digestion. Associations of mean adipocyte size with body mass index (BMI), glucose, HbA1C, blood lipids as well as mature adipocyte mitochondrial respiration were investigated. RESULTS All adipocyte area estimates derived from adipose tissue histology were not normally distributed, but rather characterized by positive skewness. The shape of the size distribution depends on the adipocyte sizing parameter and on the method used to determine adipocyte size. Despite different distribution shapes histology-derived adipocyte area, diameter, volume, and surface area consistently showed positive correlations with BMI. Furthermore, associations between adipocyte sizing parameters and glucose, HbA1C, or HDL specifically in the visceral adipose depot were revealed. Increasing subcutaneous adipocyte diameter was negatively correlated with adipocyte mitochondrial respiration. CONCLUSIONS Despite different underlying size distributions, the correlation with obesity-related traits was consistent across adipocyte sizing parameters. Decreased mitochondrial respiratory capacity with increasing subcutaneous adipocyte diameter could display a novel link between adipocyte hypertrophy and adipose tissue function.
Collapse
|
27
|
Eriksen F, Carlsson ER, Munk JK, Madsbad S, Fenger M. Fractionated free fatty acids and their relation to diabetes status after Roux-en-Y gastric bypass: A cohort study. Physiol Rep 2021; 9:e14708. [PMID: 33463892 PMCID: PMC7814490 DOI: 10.14814/phy2.14708] [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] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 01/15/2023] Open
Abstract
Bariatric surgery is associated with near-immediate remission of type 2 diabetes and recently suggested as a treatment for type 2 diabetes. Specifically, Roux-en-Y gastric bypass has been a focus of much research, but still, the mechanisms of action are only partly elucidated. We aim to investigate whether some mechanisms might be mediated by free fatty acids (FFAs). We measured eight fractionated FFAs before and up to 2 years after Roux-en-Y gastric bypass surgery in 207 patients, divided into three groups. One non-diabetic group, one diabetic group with post-operative remission and one diabetic group with persistent diabetes after surgery. Pre- and postoperative levels of fractionated FFAs were compared within and between groups. The sum of the measured FFAs were lower in the group with persistent diabetes, compared to the other groups. The pre-surgery level of linoleic acid in the group with persistent diabetes was significantly lower compared to the other two groups. The levels of fractionated FFAs decreased from pre-surgery to three months after surgery, except for oleic acid and arachidonic acid and for Docosahexaenoic acid (DHA) in the non-diabetic group. The FFAs with decreasing levels from pre-surgery to three months post-surgery are all precursors to oleic acid, arachidonic acid, and DHA, respectively, which may imply a drift, indicating that they need to be sustained at an acceptable level for optimal metabolic function. The fact that the sum of the measured FFAs is lower in the group with persistent diabetes may suggest that this group and the group with diabetes remission represent two distinct types of type 2 diabetes. It is proposed that linoleic acid could be used as a biomarker to determine the plausibility for type 2 diabetes remission after Roux-en-Y gastric bypass surgery.
Collapse
Affiliation(s)
- Freja Eriksen
- Department of Clinical BiochemistryCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Elin R. Carlsson
- Department of Clinical BiochemistryCopenhagen University Hospital HvidovreHvidovreDenmark
- Department of Clinical BiochemistryNordsjaellands HospitalUniversity of CopenhagenHillerodDenmark
| | - Jens K. Munk
- Department of Clinical BiochemistryCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Sten Madsbad
- Department of EndocrinologyCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Mogens Fenger
- Department of Clinical BiochemistryCopenhagen University Hospital HvidovreHvidovreDenmark
| |
Collapse
|
28
|
Liu F, He J, Wang H, Zhu D, Bi Y. Adipose Morphology: a Critical Factor in Regulation of Human Metabolic Diseases and Adipose Tissue Dysfunction. Obes Surg 2020; 30:5086-5100. [PMID: 33021706 PMCID: PMC7719100 DOI: 10.1007/s11695-020-04983-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/12/2020] [Accepted: 09/15/2020] [Indexed: 12/30/2022]
Abstract
Emerging evidence highlights that dysfunction of adipose tissue contributes to impaired insulin sensitivity and systemic metabolic deterioration in obese state. Of note, adipocyte hypertrophy serves as a critical event which associates closely with adipose dysfunction. An increase in cell size exacerbates hypoxia and inflammation as well as excessive collagen deposition, finally leading to metabolic dysregulation. Specific mechanisms of adipocyte hypertrophy include dysregulated differentiation and maturation of preadipocytes, enlargement of lipid droplets, and abnormal adipocyte osmolarity sensors. Also, weight loss therapies exert profound influence on adipocyte size. Here, we summarize the critical role of adipocyte hypertrophy in the development of metabolic disturbances. Future studies are required to establish a standard criterion of size measurement to better clarify the impact of adipocyte hypertrophy on changes in metabolic homeostasis.
Collapse
Affiliation(s)
- Fangcen Liu
- Department of Endocrinology, Nanjing Drum Tower Hospital Clinical College, Nanjing Medical University, Nanjing, China
| | - Jielei He
- Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hongdong Wang
- Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Dalong Zhu
- Department of Endocrinology, Nanjing Drum Tower Hospital Clinical College, Nanjing Medical University, Nanjing, China
- Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yan Bi
- Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
29
|
Mejhert N, Rydén M. Novel aspects on the role of white adipose tissue in type 2 diabetes. Curr Opin Pharmacol 2020; 55:47-52. [PMID: 33120170 DOI: 10.1016/j.coph.2020.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/09/2020] [Accepted: 09/23/2020] [Indexed: 01/14/2023]
Abstract
White adipose tissue (WAT) is a highly dynamic organ that can vary considerably in mass depending on energy balance. Data from recent cross-sectional and prospective clinical studies have revealed a set of mechanisms that link WAT dysfunction to type 2 diabetes. This review focuses on three of the most important pathophysiological processes that distinguish WAT in the insulin resistant state: regional WAT distribution, adipocyte hypertrophy and lipid turnover. Together, these disturbances attenuate the lipid storage capacity of WAT leading to ectopic fat deposition in peripheral tissues such as skeletal muscle, liver and vessels ultimately leading to type 2 diabetes and cardiovascular complications. The possible approaches to therapeutically target dysfunctional WAT are also discussed.
Collapse
Affiliation(s)
- Niklas Mejhert
- Department of Medicine (H7), Karolinska Institutet, C2-94, Karolinska University Hospital, 141 86, Stockholm, Sweden.
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, C2-94, Karolinska University Hospital, 141 86, Stockholm, Sweden.
| |
Collapse
|
30
|
Kerr AG, Andersson DP, Rydén M, Arner P, Dahlman I. Long-term changes in adipose tissue gene expression following bariatric surgery. J Intern Med 2020; 288:219-233. [PMID: 32406570 DOI: 10.1111/joim.13066] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Patients undergoing bariatric surgery present long-term metabolic improvements and reduced type 2 diabetes risk, despite long-term weight regain. We hypothesized that part of these protective effects could be linked to altered gene expression in white adipose tissue (WAT). METHODS Transcriptomic profiling by gene microarray was performed in abdominal subcutaneous WAT from women before (n = 50) and two (n = 49) and five (n = 38) years after Roux-en-Y gastric bypass (RYGB) surgery as well as in 28 age-matched nonoperated women. RESULTS In the obese women, the average body weight decrease was 38 kg 2 years postsurgery followed by an 8 kg weight regain between 2 and 5 years. Most of the long-term changes in WAT gene expression occurred during the first 2 years. However, a subset of genes encoding proteins involved in inflammation displayed a continued decrease between baseline, 2 and 5 years, respectively; that is an expression pattern independent of body weight regain. Expression of 71 of these genes correlated with measurements of adipocyte morphology or serum adipokine levels. CONCLUSION The continuous improvement in WAT inflammatory gene expression, despite body weight relapse, may contribute to the sustained effects on adipose morphology after bariatric surgery.
Collapse
Affiliation(s)
- A G Kerr
- From the , Lipid Laboratory, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - D P Andersson
- From the , Lipid Laboratory, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - M Rydén
- From the , Lipid Laboratory, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - P Arner
- From the , Lipid Laboratory, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - I Dahlman
- From the , Lipid Laboratory, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Metabolic adaptations after bariatric surgery: adipokines, myokines and hepatokines. Curr Opin Pharmacol 2020; 52:67-74. [PMID: 32688292 DOI: 10.1016/j.coph.2020.06.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/23/2022]
Abstract
This review addresses the impact of bariatric surgery on the endocrine aspects of white adipose tissue, muscle and the liver. We describe literature supporting the notion that adipokines, myokines and hepatokines likely act in concert and drive many of the long-term metabolic improvements following surgery. Circulating adiponectin is increased while secretion of pro-inflammatory interleukins (1, 6 and 8) decreases, alongside leptin secretion. The metabolic improvements observed in the muscle might relate to reduction of myokines contributing to insulin resistance (including myostatin, brain-derived neurotrophic factor and fibroblast growth factor-21). Subject to exception, hepatokine secretion is generally increased (such as insulin-like growth factor-binding protein 2, adropin and sex hormone-binding globulin). In conclusion, bariatric surgery restores metabolic functions by enhancing the time-dependent secretion of anti-inflammatory, insulin-sensitizing and antilipemic factors. Further research is needed to understand the molecular mechanisms by which these factors may trigger the remission of obesity-related comorbidities following bariatric surgery.
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW To give an updated review on the underlying mechanisms and clinical effects of improved glucose control after bariatric surgery. RECENT FINDINGS The basic principles of the mechanism for the metabolic effects of bariatric surgery can be categorized into calorie restriction, deviation of nutrients, and reduced amounts of adipose tissue. Recent findings suggest the importance of early changes following deviation of nutrients to more distal parts of the small bowel resulting in altered release of gastrointestinal hormones, altered gut microbiota, and weight-reduction. In the long-term, loss of adipose tissue results in reduced inflammation and improved insulin sensitivity. From a clinical perspective these changes are associated with remission of diabetes in patients with morbid obesity and type 2 diabetes, prevention of diabetes in patients with insulin resistance without overt type 2 diabetes and prevention of both microvascular and macrovascular complications for all patients with morbid obesity. SUMMARY At present, bariatric surgery remains the most effective treatment option to improve glucose control and long-term complications associated with hyperglycemia in patients with obesity.Although the mechanisms behind these metabolic effects remain only partially understood, further knowledge on these complex mechanisms may help identifying durable treatment options for morbid obesity and important metabolic comorbidities.
Collapse
Affiliation(s)
- Erik Stenberg
- Faculty of Medicine and Health, Örebro University & Department of Surgery, Region Örebro County, Örebro, Sweden
| | - Anders Thorell
- Department of Clinical Science, Danderyds Hospital, Karolinska Institutet
- Department of Surgery, Ersta hospital, Stockholm, Sweden
| |
Collapse
|
33
|
Wu KKL, Cheung SWM, Cheng KKY. NLRP3 Inflammasome Activation in Adipose Tissues and Its Implications on Metabolic Diseases. Int J Mol Sci 2020; 21:E4184. [PMID: 32545355 PMCID: PMC7312293 DOI: 10.3390/ijms21114184] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
Adipose tissue is an active endocrine and immune organ that controls systemic immunometabolism via multiple pathways. Diverse immune cell populations reside in adipose tissue, and their composition and immune responses vary with nutritional and environmental conditions. Adipose tissue dysfunction, characterized by sterile low-grade chronic inflammation and excessive immune cell infiltration, is a hallmark of obesity, as well as an important link to cardiometabolic diseases. Amongst the pro-inflammatory factors secreted by the dysfunctional adipose tissue, interleukin (IL)-1β, induced by the NLR family pyrin domain-containing 3 (NLRP3) inflammasome, not only impairs peripheral insulin sensitivity, but it also interferes with the endocrine and immune functions of adipose tissue in a paracrine manner. Human studies indicated that NLRP3 activity in adipose tissues positively correlates with obesity and its metabolic complications, and treatment with the IL-1β antibody improves glycaemia control in type 2 diabetic patients. In mouse models, genetic or pharmacological inhibition of NLRP3 activation pathways or IL-1β prevents adipose tissue dysfunction, including inflammation, fibrosis, defective lipid handling and adipogenesis, which in turn alleviates obesity and its related metabolic disorders. In this review, we summarize both the negative and positive regulators of NLRP3 inflammasome activation, and its pathophysiological consequences on immunometabolism. We also discuss the potential therapeutic approaches to targeting adipose tissue inflammasome for the treatment of obesity and its related metabolic disorders.
Collapse
Affiliation(s)
| | | | - Kenneth King-Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (K.K.-L.W.); (S.W.-M.C.)
| |
Collapse
|
34
|
Usefulness of surrogate markers to determine insulin action in fat cells. Int J Obes (Lond) 2020; 44:2436-2443. [PMID: 32398753 DOI: 10.1038/s41366-020-0592-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/02/2020] [Accepted: 04/29/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Obesity is a major factor behind insulin resistance. The validity of simple biochemical surrogate measures to estimate insulin resistance at the fat cell level is unclear. OBJECTIVE To investigate if the surrogate measures HOMA-IR (glucose/insulin product) and Adipo-IR (fatty acids/insulin product) reflect insulin action on glucose/lipid metabolism in fat cells. DESIGN Insulin-induced lipogenesis and lipolysis inhibition (antilipolysis) in subcutaneous fat cells were investigated for sensitivity (reflecting receptor-near events) and responsiveness (i.e., maximum action reflecting distal post-receptor events) in 363 subjects. Results were compared with log10 transformed values for HOMA-IR and Adipo-IR. RESULTS Individually, the four measures of in vitro insulin action on fat cells correlated significantly (p < 0.0001) but weakly with each other (adjusted r2 0.05-0.23). HOMA-IR and Adipo-IR correlated strongly with each other (adjusted r2 = 0.81). Using Spearman or simple linear regression all in vitro measures except antilipolytic responsiveness expressed per lipid weight, correlated significantly with Adipo-IR or HOMA-IR (p values <0.0001). Similar relationships remained after combined correction for age, body mass index and sex. Together, the four in vitro measures explained 50% of the variability in HOMA-IR and ADIPO-IR (p < 0.0001). Receiver-operating characteristic analysis showed good sensitivity and specificity for Adipo-IR and HOMA-IR to detect combined insulin resistance of antilipolysis and lipogenesis in fat cells (area under the curve = 0.8). CONCLUSIONS Insulin action at the receptor and post-receptor levels on lipolysis and lipogenesis in fat cells correlates significantly with Adipo-IR and HOMA-IR. Both surrogate measures give similar information about insulin resistance of glucose and lipid metabolism in fat cells.
Collapse
|
35
|
Eckel-Mahan K, Ribas Latre A, Kolonin MG. Adipose Stromal Cell Expansion and Exhaustion: Mechanisms and Consequences. Cells 2020; 9:cells9040863. [PMID: 32252348 PMCID: PMC7226766 DOI: 10.3390/cells9040863] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue (AT) is comprised of a diverse number of cell types, including adipocytes, stromal cells, endothelial cells, and infiltrating leukocytes. Adipose stromal cells (ASCs) are a mixed population containing adipose progenitor cells (APCs) as well as fibro-inflammatory precursors and cells supporting the vasculature. There is growing evidence that the ability of ASCs to renew and undergo adipogenesis into new, healthy adipocytes is a hallmark of healthy fat, preventing disease-inducing adipocyte hypertrophy and the spillover of lipids into other organs, such as the liver and muscles. However, there is building evidence indicating that the ability for ASCs to self-renew is not infinite. With rates of ASC proliferation and adipogenesis tightly controlled by diet and the circadian clock, the capacity to maintain healthy AT via the generation of new, healthy adipocytes appears to be tightly regulated. Here, we review the contributions of ASCs to the maintenance of distinct adipocyte pools as well as pathogenic fibroblasts in cancer and fibrosis. We also discuss aging and diet-induced obesity as factors that might lead to ASC senescence, and the consequences for metabolic health.
Collapse
Affiliation(s)
- Kristin Eckel-Mahan
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, TX 77030, USA;
| | - Aleix Ribas Latre
- Helmholtz Institute for Metabolic, Obesity and Vascular Research Center, D-04103 Leipzig, Germany;
| | - Mikhail G. Kolonin
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, TX 77030, USA;
- Correspondence:
| |
Collapse
|
36
|
Eriksson Hogling D, Bäckdahl J, Thorell A, Rydén M, Andersson DP. Metabolic Impact of Body Fat Percentage Independent of Body Mass Index in Women with Obesity Remission After Gastric Bypass. Obes Surg 2020; 30:1086-1092. [PMID: 31832934 PMCID: PMC7347509 DOI: 10.1007/s11695-019-04304-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND/OBJECTIVE Body mass index (BMI) is central when evaluating treatment effect after gastric bypass. The metabolic impact of BMI-independent differences in body fat percentage (BF%) after gastric bypass is not fully understood. We compared metabolic and adipose tissue characteristics in women with high versus low BF% independent of BMI after obesity remission following gastric bypass. SUBJECTS/METHODS A cohort of 215 women was included at baseline. A total of 166 women were re-examined 2 years after gastric bypass, whereof 130 had obesity remission (BMI < 30 kg/m2). Anthropometric parameters, blood pressure, and lipids were measured. Total and regional body fat mass was determined by dual-energy X-ray absorptiometry. Insulin sensitivity was assessed by homeostasis model assessment of insulin resistance (HOMA-IR) and hyperinsulinemic euglycemic clamp (M value). Adipocyte size and number were determined. RESULTS Of the 130 women with obesity remission, 64 had BF% ≥ 35 and 65 < 35. Independent of BMI, high BF% were associated with higher HOMA-IR (P = 0.021), lower M value (P = 0.0046), higher triglycerides (P = 0.013), higher visceral/total and android/gynoid fat mass ratios (P = 0.0032 and 0.0003 respectively), and larger subcutaneous fat cell volume (P < 0.0001) 2 years after gastric bypass. No differences in anthropometric measures, glucose, blood pressure, or fat cell number were observed. CONCLUSIONS Independent of BMI, patients with higher BF% displayed lower insulin sensitivity, higher triglyceride levels, central fat distribution, and larger subcutaneous adipocytes 2 years after gastric bypass. Thus, determination of BF% provides additional information of metabolic characteristics at follow-up of non-obese patients after gastric bypass.
Collapse
Affiliation(s)
- Daniel Eriksson Hogling
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | - Jesper Bäckdahl
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | - Anders Thorell
- Department of Clinical Sciences, Danderyd Hospital & Department of Surgery, Ersta Hospital, Karolinska Institutet, 116 91 Stockholm, Sweden
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | - Daniel P. Andersson
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| |
Collapse
|
37
|
Varela-Rodríguez BM, Juiz-Valiña P, Varela L, Outeiriño-Blanco E, Bravo SB, García-Brao MJ, Mena E, Noguera JF, Valero-Gasalla J, Cordido F, Sangiao-Alvarellos S. Beneficial Effects of Bariatric Surgery-Induced by Weight Loss on the Proteome of Abdominal Subcutaneous Adipose Tissue. J Clin Med 2020; 9:jcm9010213. [PMID: 31941045 PMCID: PMC7019912 DOI: 10.3390/jcm9010213] [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: 11/27/2019] [Revised: 12/20/2019] [Accepted: 01/08/2020] [Indexed: 12/16/2022] Open
Abstract
Bariatric surgery (BS) is the most effective treatment for obesity and has a positive impact on cardiometabolic risk and in the remission of type 2 diabetes. Following BS, the majority of fat mass is lost from the subcutaneous adipose tissue depot (SAT). However, the changes in this depot and functions and as well as its relative contribution to the beneficial effects of this surgery are still controversial. With the aim of studying altered proteins and molecular pathways in abdominal SAT (aSAT) after body weight normalization induced by BS, we carried out a proteomic approach sequential window acquisition of all theoretical mass spectra (SWATH-MS) analysis. These results were complemented by Western blot, electron microscopy and RT-qPCR. With all of the working tools mentioned, we confirmed that after BS, up-regulated proteins were associated with metabolism, the citric acid cycle and respiratory electron transport, triglyceride catabolism and metabolism, formation of ATP, pyruvate metabolism, glycolysis/gluconeogenesis and thermogenesis among others. In contrast, proteins with decreased values are part of the biological pathways related to the immune system. We also confirmed that obesity caused a significant decrease in mitochondrial density and coverage, which was corrected by BS. Together, these findings reveal specific molecular mechanisms, genes and proteins that improve adipose tissue function after BS characterized by lower inflammation, increased glucose uptake, higher insulin sensitivity, higher de novo lipogenesis, increased mitochondrial function and decreased adipocyte size.
Collapse
Affiliation(s)
- Bárbara María Varela-Rodríguez
- Endocrine, Nutritional and Metabolic Diseases Group, Faculty of Health Sciences, Universidade da Coruña, Campus de Oza, 15006 A Coruña, Spain; (B.M.V.-R.); (P.J.-V.); (F.C.)
- INIBIC (Instituto de Investigación Biomédica de A Coruña), Xubias de Arriba, 84. 15006 A Coruña, Spain
- CICA (Centro de Investigaciones Científicas Avanzadas), As Carballeiras, s/n Campus de, San Vicente de Elviña, 15008 A Coruña, Spain
| | - Paula Juiz-Valiña
- Endocrine, Nutritional and Metabolic Diseases Group, Faculty of Health Sciences, Universidade da Coruña, Campus de Oza, 15006 A Coruña, Spain; (B.M.V.-R.); (P.J.-V.); (F.C.)
- INIBIC (Instituto de Investigación Biomédica de A Coruña), Xubias de Arriba, 84. 15006 A Coruña, Spain
- CICA (Centro de Investigaciones Científicas Avanzadas), As Carballeiras, s/n Campus de, San Vicente de Elviña, 15008 A Coruña, Spain
| | - Luis Varela
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA;
| | - Elena Outeiriño-Blanco
- Department of Endocrinology, Hospital Universitario A Coruña, A Coruña, 15006 A Coruña, Spain;
| | - Susana Belén Bravo
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15705 A Coruña, Spain;
| | - María Jesús García-Brao
- Department of Digestive and General Surgery, Hospital Universitario A Coruña, 15006 A Coruña, Spain; (M.J.G.-B.); (E.M.); (J.F.N.)
| | - Enrique Mena
- Department of Digestive and General Surgery, Hospital Universitario A Coruña, 15006 A Coruña, Spain; (M.J.G.-B.); (E.M.); (J.F.N.)
| | - José Francisco Noguera
- Department of Digestive and General Surgery, Hospital Universitario A Coruña, 15006 A Coruña, Spain; (M.J.G.-B.); (E.M.); (J.F.N.)
| | - Javier Valero-Gasalla
- Department of Plastic, Reconstructive & Aesthetic Surgery. Hospital Universitario A Coruña, 15006 A Coruña, Spain;
| | - Fernando Cordido
- Endocrine, Nutritional and Metabolic Diseases Group, Faculty of Health Sciences, Universidade da Coruña, Campus de Oza, 15006 A Coruña, Spain; (B.M.V.-R.); (P.J.-V.); (F.C.)
- INIBIC (Instituto de Investigación Biomédica de A Coruña), Xubias de Arriba, 84. 15006 A Coruña, Spain
- CICA (Centro de Investigaciones Científicas Avanzadas), As Carballeiras, s/n Campus de, San Vicente de Elviña, 15008 A Coruña, Spain
- Department of Endocrinology, Hospital Universitario A Coruña, A Coruña, 15006 A Coruña, Spain;
| | - Susana Sangiao-Alvarellos
- Endocrine, Nutritional and Metabolic Diseases Group, Faculty of Health Sciences, Universidade da Coruña, Campus de Oza, 15006 A Coruña, Spain; (B.M.V.-R.); (P.J.-V.); (F.C.)
- INIBIC (Instituto de Investigación Biomédica de A Coruña), Xubias de Arriba, 84. 15006 A Coruña, Spain
- CICA (Centro de Investigaciones Científicas Avanzadas), As Carballeiras, s/n Campus de, San Vicente de Elviña, 15008 A Coruña, Spain
- Correspondence:
| |
Collapse
|
38
|
Abstract
Accumulating knowledge on the biology and function of the adipose tissue has led to a major shift in our understanding of its role in health and disease. The adipose tissue is now recognized as a crucial regulator of cardiovascular health, mediated by the secretion of several bioactive products, including adipocytokines, microvesicles and gaseous messengers, with a wide range of endocrine and paracrine effects on the cardiovascular system. The adipose tissue function and secretome are tightly controlled by complex homeostatic mechanisms and local cell-cell interactions, which can become dysregulated in obesity. Systemic or local inflammation and insulin resistance lead to a shift in the adipose tissue secretome from anti-inflammatory and anti-atherogenic towards a pro-inflammatory and pro-atherogenic profile. Moreover, the interplay between the adipose tissue and the cardiovascular system is bidirectional, with vascular-derived and heart-derived signals directly affecting adipose tissue biology. In this Review, we summarize the current knowledge of the biology and regional variability of adipose tissue in humans, deciphering the complex molecular mechanisms controlling the crosstalk between the adipose tissue and the cardiovascular system, and their possible clinical translation. In addition, we highlight the latest developments in adipose tissue imaging for cardiovascular risk stratification and discuss how therapeutic targeting of the adipose tissue can improve prevention and treatment of cardiovascular disease.
Collapse
|
39
|
Adipose lipid turnover and long-term changes in body weight. Nat Med 2019; 25:1385-1389. [DOI: 10.1038/s41591-019-0565-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 07/30/2019] [Indexed: 01/08/2023]
|
40
|
Zhang C, Zhang J, Liu W, Chen X, Liu Z, Zhou Z. Improvements in humoral immune function and glucolipid metabolism after laparoscopic sleeve gastrectomy in patients with obesity. Surg Obes Relat Dis 2019; 15:1455-1463. [DOI: 10.1016/j.soard.2019.05.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/30/2019] [Accepted: 05/12/2019] [Indexed: 02/08/2023]
|
41
|
Rydén M, Petrus P, Andersson DP, Medina-Gómez G, Escasany E, Corrales Cordón P, Dahlman I, Kulyté A, Arner P. Insulin action is severely impaired in adipocytes of apparently healthy overweight and obese subjects. J Intern Med 2019; 285:578-588. [PMID: 30758089 DOI: 10.1111/joim.12887] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Many overweight/obese subjects appear metabolically healthy with normal in vivo insulin sensitivity. Still, they have increased long-term risk of developing type 2 diabetes. We hypothesized that adipose tissue dysfunction involving decreased insulin action in adipocytes is present in apparently healthy overweight/obese subjects. DESIGN/METHODS Subjects with normal metabolic health according to Adult Treatment Panel-III or Framingham risk score criteria were subdivided into 67 lean, 32 overweight and 37 obese according to body mass index. They were compared with 200 obese individuals with metabolic syndrome. Insulin sensitivity and maximum action on inhibition of lipolysis and stimulation of lipogenesis was determined in subcutaneous adipocytes. Gene expression was determined by micro-array and qPCR. DNA methylation was assessed by array, pyrosequencing and reporter assays. RESULTS Compared with lean, adipocytes in overweight/obese displayed marked reductions in insulin sensitivity in both antilipolysis and lipogenesis as well as an attenuated maximum lipogenic response. Among these, only antilipolysis sensitivity correlated with whole-body insulin sensitivity. These differences were already evident in the overweight state, were only slightly worse in the unhealthy obese state and were not related to fat cell size. Adipose tissue analyses linked this to reduced expression of the insulin signalling protein AKT2, which associated with increased methylation at regulatory sites in the AKT2 promoter. CONCLUSIONS Apparently healthy subjects have severely disturbed adipocyte insulin signalling already in the overweight state which involves epigenetic dysregulation of AKT2. This may constitute an early defect in insulin action that appears even upon modest increases in fat mass.
Collapse
Affiliation(s)
- M Rydén
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - P Petrus
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - D P Andersson
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - G Medina-Gómez
- Departamento de Ciencias Básicas de la Salud, Área Bioquímica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Spain
| | - E Escasany
- Departamento de Ciencias Básicas de la Salud, Área Bioquímica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Spain
| | - P Corrales Cordón
- Departamento de Ciencias Básicas de la Salud, Área Bioquímica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Spain
| | - I Dahlman
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - A Kulyté
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - P Arner
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
42
|
Kerr AG, Sinha I, Dadvar S, Arner P, Dahlman I. Epigenetic regulation of diabetogenic adipose morphology. Mol Metab 2019; 25:159-167. [PMID: 31031182 PMCID: PMC6600120 DOI: 10.1016/j.molmet.2019.04.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 04/13/2019] [Indexed: 12/17/2022] Open
Abstract
Objective Hypertrophic white adipose tissue (WAT) morphology is associated with insulin resistance and type 2 diabetes. The mechanisms governing hyperplastic versus hypertrophic WAT expansion are poorly understood. We assessed if epigenetic modifications in adipocytes are associated with hypertrophic adipose morphology. A subset of genes with differentially methylated CpG-sites (DMS) in the promoters was taken forward for functional evaluation. Methods The study included 126 women who underwent abdominal subcutaneous biopsy to determine adipose morphology. Global transcriptome profiling was performed on WAT from 113 of the women, and CpG methylome profiling on isolated adipocytes from 78 women. Small interfering RNAs (siRNA) knockdown in human mesenchymal stem cells (hMSCs) was used to assess influence of specific genes on lipid storage. Results A higher proportion of CpG-sites were methylated in hypertrophic compared to hyperplastic WAT. Methylation at 35,138 CpG-sites was found to correlate to adipose morphology. 2,102 of these CpG-sites were also differentially methylated in T2D; 98% showed directionally consistent change in methylation in WAT hypertrophy and T2D. We identified 2,508 DMS in 638 adipose morphology-associated genes where methylation correlated with gene expression. These genes were over-represented in gene sets relevant to WAT hypertrophy, such as insulin resistance, lipolysis, extracellular matrix organization, and innate immunity. siRNA knockdown of ADH1B, AZGP1, C14orf180, GYG2, HADH, PRKAR2B, PFKFB3, and AQP7 influenced lipid storage and metabolism. Conclusion CpG methylation could be influential in determining adipose morphology and thereby constitute a novel antidiabetic target. We identified C14orf180 as a novel regulator of adipocyte lipid storage and possibly differentiation. Hypertrophic adipose morphology display a distinct adipocyte CpG-methylome profile. Adipose hypertrophy and type 2 diabetes display strong overlap in CpG-methylome profile. C14orf180 is a novel regulator of adipocyte lipid storage and possibly adipogenesis.
Collapse
Affiliation(s)
- A G Kerr
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - I Sinha
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - S Dadvar
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - P Arner
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - I Dahlman
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
43
|
Zhou K, Wolski K, Malin SK, Aminian A, Schauer PR, Bhatt DL, Kashyap SR. IMPACT OF WEIGHT LOSS TRAJECTORY FOLLOWING RANDOMIZATION TO BARIATRIC SURGERY ON LONG-TERM DIABETES GLYCEMIC AND CARDIOMETABOLIC PARAMETERS. Endocr Pract 2019; 25:572-579. [PMID: 30865529 DOI: 10.4158/ep-2018-0522] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Objective: It is unclear whether acute weight loss or the chronic trajectory of weight loss after bariatric surgery is associated with long-term type 2 diabetes mellitus (T2DM) glycemic improvement. This ancillary study of the Surgical Treatment and Medications Potentially Eradicate Diabetes Efficiently (STAMPEDE) trial aimed to answer this question. Methods: In STAMPEDE, 150 patients with T2DM were randomized to bariatric surgery, and 96 had 5-year follow-up. Data post-Roux-en-Y gastric bypass (RYGB, n = 49) and sleeve gastrectomy (SG, n = 47) were analyzed. We defined percent weight loss in the first year as negative percent decrease from baseline weight to lowest weight in the first year. Percent weight regain was positive percent change from lowest weight in the first year to fifth year. Weight change was then correlated with cardiometabolic (CM) and glycemic outcomes at 5 years using Spearman rank correlations and multivariate analysis. Results: In both RYGB and SG, less weight loss in the first year positively correlated with higher 5-year glycated hemoglobin (HbA1c) (RYGB, β = +0.13; P<.001 and SG, β = 0.14; P<.001). In SG, greater weight regain from nadir positively correlated with higher HbA1c (β = 0.06; P = .02), but not in RYGB. Reduced first-year weight loss was also correlated with increased 5-year triglycerides (β = 1.81; P = .01), but not systolic blood pressure. Weight regain did not correlate with CM outcomes. Conclusion: Acute weight loss may be more important for T2DM glycemic control following both RYGB and SG as compared with weight regain. Clinicians should aim to assist patients with achieving maximal weight loss in the first year post-op to maximize long-term health of patients. Abbreviations: BMI = body mass index; HbA1c = glycated hemoglobin; RYGB = Roux-en-Y gastric bypass; SBP = systolic blood pressure; SG = sleeve gastrectomy; STAMPEDE = Surgical Treatment and Medications Potentially Eradicate Diabetes Efficiently; T2DM = type 2 diabetes mellitus; TG = triglyceride.
Collapse
|
44
|
Courcoulas AP, Stefater MA, Shirley E, Gourash WF, Stylopoulos N. The Feasibility of Examining the Effects of Gastric Bypass Surgery on Intestinal Metabolism: Prospective, Longitudinal Mechanistic Clinical Trial. JMIR Res Protoc 2019; 8:e12459. [PMID: 30679147 PMCID: PMC6483060 DOI: 10.2196/12459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/05/2018] [Accepted: 12/09/2018] [Indexed: 01/01/2023] Open
Abstract
Background Bariatric surgery, especially Roux-en-Y gastric bypass (RYGB), is the best treatment for severe obesity and its complications including type 2 diabetes mellitus (T2DM). Understanding the mechanisms responsible for the beneficial metabolic effects will help to engineer ways to improve the procedure or produce these effects without surgery. Objective The aim is to present data on recruitment and feasibility of a translational study designed to collect intestinal samples before and after bariatric surgery. The goal of biobanking is to allow future studies to test the hypothesis that the mechanism of action of RYGB involves specific changes in the postsurgical short- and long-term metabolism and morphology of the jejunum (Roux limb). Specifically, to test whether the intestine enhances its metabolism and activity after RYGB and increases its fuel utilization, we designed a prospective, longitudinal study, which involved the recruitment of candidates for RYGB with and without T2DM. We describe the tissue bank that we have generated, and our experience, hoping to further facilitate the performance of longitudinal mechanistic studies in human patients undergoing bariatric surgery and especially those involving post-RYGB intestinal biology. Methods We conducted a trial to characterize the effects of RYGB on intestinal metabolism. Intestinal tissue samples were collected from the jejunum at surgery, 1, 6, and 12 months postoperatively for the analysis of intestinal gene expression and metabolomic and morphologic changes. The target number of patients who completed at least the 6-month follow-up was 26, and we included a 20% attrition rate, increasing the total number to 32. Results To enroll 26 patients, we had to approach 79 potential participants. A total of 37 agreed to participate and started the study; 33, 30, and 26 active participants completed their 1-month, 6-month, and 12-month studies, respectively. Three participants withdrew, and 30 participants are still active. Altruism and interest in research were the most common reasons for participation. Important factors for feasibility and successful retention included (1) large volume case flow, (2) inclusion and exclusion criteria broad enough to capture a large segment of the patient population but narrow enough to ensure the completion of study aims and protection of safety concerns, (3) accurate assessment of willingness and motivation to participate in a study, (4) seamless integration of the recruitment process into normal clinical flow, (5) financial reimbursement and nonfinancial rewards and gestures of appreciation, and (6) nonburdensome follow-up visits and measures and reasonable time allotted. Conclusions Human translational studies of the intestinal mechanisms of metabolic and weight changes after bariatric surgery are important and feasible. A tissue bank with unique samples has been established that could be used by investigators in many research fields, further enabling mechanistic studies on the effects of bariatric surgery. Trial Registration ClinicalTrials.gov NCT02710370; https://clinicaltrials.gov/ct2/show/NCT02710370 (Archived by WebCite at http://www.webcitation.org/75HrQT8Dl)
Collapse
Affiliation(s)
| | | | - Eleanor Shirley
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - William F Gourash
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | | |
Collapse
|
45
|
Vreeken D, Wiesmann M, Deden LN, Arnoldussen IAC, Aarts E, Kessels RPC, Kleemann R, Hazebroek EJ, Aarts EO, Kiliaan AJ. Study rationale and protocol of the BARICO study: a longitudinal, prospective, observational study to evaluate the effects of weight loss on brain function and structure after bariatric surgery. BMJ Open 2019; 9:e025464. [PMID: 30782752 PMCID: PMC6340014 DOI: 10.1136/bmjopen-2018-025464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Weight loss after bariatric surgery (BS) is often associated with improved cognition and structural brain recovery. However, improved cognition after BS is not always exhibited by patients, in fact, in some cases there is even a decline in cognition. Long-term consequences of BS weight loss, in terms of obesity and related diseases, can be hard to determine due to studies having short follow-up periods and small sample sizes.The aim of the BARICO study (BAriatric surgery Rijnstate and Radboudumc neuroImaging and Cognition in Obesity) is to determine the long-term effect of weight loss after BS on brain function and structure, using sensitive neuropsychological tests and (functional) MRI ((f)MRI). Secondary study endpoints are associated with changes in metabolic and inflammation status of adipose tissue, liver and gut, in relation to brain structure and function. Also, the possible correlation between weight loss, gut microbiota composition change and neuropsychological outcomes will be investigated. METHODS AND ANALYSIS Data from 150 Dutch BS patients (ages between 35 and 55, men and women) will be collected at various time points between 2 months before and up to 10 years after surgery. Neuropsychological tests, questionnaires, blood, faeces and tissue samples will be collected before, during and after surgery to measure changes in cognition, microbiota, metabolic activity and inflammation over time. A subgroup of 75 participants will undergo (f)MRI in relation to executive functioning (determined by the Stroop task), grey and white matter volumes and cerebral blood flow. Regression analyses will be used to explore associations between weight loss and outcome measures. ETHICS AND DISSEMINATION This study has been approved by the medical review ethics committee CMO Region Arnhem and Nijmegen (NL63493.091.17). Research findings will be published in peer-reviewed journals and at conferences. TRIAL REGISTRATION NUMBER NTR7288.
Collapse
Affiliation(s)
- Debby Vreeken
- Department of Surgery, Rijnstate Hospital, Vitalys Clinic, Arnhem, The Netherlands
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maximilian Wiesmann
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Laura N Deden
- Department of Surgery, Rijnstate Hospital, Vitalys Clinic, Arnhem, The Netherlands
| | - Ilse A C Arnoldussen
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Esther Aarts
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Roy P C Kessels
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Medical Psychology, Radboud University Medical Center, Nijmegen, The Netherlands
- Vincent van Gogh Institute for Psychiatry, Venray, The Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Eric J Hazebroek
- Department of Surgery, Rijnstate Hospital, Vitalys Clinic, Arnhem, The Netherlands
| | - Edo O Aarts
- Department of Surgery, Rijnstate Hospital, Vitalys Clinic, Arnhem, The Netherlands
| | - Amanda J Kiliaan
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
46
|
Andersson DP, Dahlman I, Eriksson Hogling D, Bäckdahl J, Toft E, Qvisth V, Näslund E, Thorell A, Rydén M, Arner P. Improved metabolism and body composition beyond normal levels following gastric bypass surgery: a longitudinal study. J Intern Med 2019; 285:92-101. [PMID: 30141528 DOI: 10.1111/joim.12824] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The cardiometabolic risk profile improves following bariatric surgery. However, the degree of improvement in relation to weight-stable control subjects is unknown. OBJECTIVES To study the differences in cardiometabolic risk profile between formerly obese patients following Roux-en-Y gastric bypass (RYGB) surgery and control subjects. METHODS Subjects undergoing RYGB and reaching a BMI <30 kg m-2 2 years postsurgery were matched with control subjects regarding age, sex and BMI. The following examinations were performed: insulin sensitivity measured by hyperinsulinaemic-euglycaemic clamp, insulin clearance, homeostatic model assessment of insulin resistance (HOMA-IR), lipid profile, inflammatory marker levels, dual-energy X-ray absorptiometry and subcutaneous adipose tissue cellularity (fat cell size and number). RESULTS Sixty-nine subjects undergoing RYGB were matched to a control subject. Insulin sensitivity measured by hyperinsulinaemic-euglycaemic clamp, blood pressure, inflammatory status and glucose, triglyceride and HDL cholesterol levels were comparable to values of control subjects. However, HOMA-IR (1.0 ± 0.5 vs. 1.3 ± 0.7, P = 0.005), insulin clearance (0.38 ± 0.08 vs. 0.34 ± 0.08 μL m-2 min-1 , P < 0.0001) and circulating levels of insulin (31 ± 15 vs. 37 ± 17 pmol L-1 , P = 0.008), total cholesterol (4.1 ± 0.7 vs. 4.8 ± 0.9 mmol L-1 , P < 0.0001) and LDL cholesterol (2.1 ± 0.6 vs. 2.9 ± 0.8 mmol L-1 , P < 0.0001) were improved beyond the levels in matched control subjects. Furthermore, formerly obese subjects had higher lean and lower fat mass as well as a more benign type of adipose cellularity (hyperplasia with many small fat cells) compared to control subjects. CONCLUSIONS Subjects who underwent RYGB and reached a postobese state demonstrated a beneficial body composition, slightly increased insulin sensitivity as indirectly measured by HOMA-IR and higher insulin clearance, lower atherogenic lipid/lipoprotein levels and benign adipocyte morphology compared with control subjects who had never been obese. In line with previous results, our findings may in part explain why RYGB confers long-term protection against metabolic complications.
Collapse
Affiliation(s)
- D P Andersson
- Department of Medicine (H7), Karolinska University Hospital, Stockholm, Sweden
| | - I Dahlman
- Department of Medicine, Ersta Hospital, Stockholm, Sweden
| | - D Eriksson Hogling
- Department of Medicine (H7), Karolinska University Hospital, Stockholm, Sweden
| | - J Bäckdahl
- Department of Medicine (H7), Karolinska University Hospital, Stockholm, Sweden
| | - E Toft
- Department of Medicine, Ersta Hospital, Stockholm, Sweden
| | - V Qvisth
- Department of Medicine, Ersta Hospital, Stockholm, Sweden
| | - E Näslund
- Department of Clinical Sciences, Danderyd Hospital, Stockholm, Sweden
| | - A Thorell
- Department of Surgery, Karolinska Institutet, Ersta Hospital, Stockholm, Sweden
| | - M Rydén
- Department of Medicine (H7), Karolinska University Hospital, Stockholm, Sweden
| | - P Arner
- Department of Medicine (H7), Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
47
|
Pérez-Pevida B, Escalada J, Miras AD, Frühbeck G. Mechanisms Underlying Type 2 Diabetes Remission After Metabolic Surgery. Front Endocrinol (Lausanne) 2019; 10:641. [PMID: 31608010 PMCID: PMC6761227 DOI: 10.3389/fendo.2019.00641] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 09/04/2019] [Indexed: 01/19/2023] Open
Abstract
Type 2 diabetes prevalence is increasing dramatically worldwide. Metabolic surgery is the most effective treatment for selected patients with diabetes and/or obesity. When compared to intensive medical therapy and lifestyle intervention, metabolic surgery has shown superiority in achieving glycemic improvement, reducing number of medications and cardiovascular risk factors, which translates in long-term benefits on cardiovascular morbidity and mortality. The mechanisms underlying diabetes improvement after metabolic surgery have not yet been clearly understood but englobe a complex interaction among improvements in beta cell function and insulin secretion, insulin sensitivity, intestinal gluconeogenesis, changes in glucose utilization, and absorption by the gut and changes in the secretory pattern and morphology of adipose tissue. These are achieved through different mediators which include an enhancement in gut hormones release, especially, glucagon-like peptide 1, changes in bile acids circulation, gut microbiome, and glucose transporters expression. Therefore, this review aims to provide a comprehensive appraisal of what is known so far to better understand the mechanisms through which metabolic surgery improves glycemic control facilitating future research in the field.
Collapse
Affiliation(s)
- Belén Pérez-Pevida
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, Hammersmith Campus, London, United Kingdom
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- *Correspondence: Belén Pérez-Pevida
| | - Javier Escalada
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), ISCIII, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Alexander D. Miras
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, Hammersmith Campus, London, United Kingdom
| | - Gema Frühbeck
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), ISCIII, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Gema Frühbeck
| |
Collapse
|
48
|
Petrus P, Mejhert N, Corrales P, Lecoutre S, Li Q, Maldonado E, Kulyté A, Lopez Y, Campbell M, Acosta JR, Laurencikiene J, Douagi I, Gao H, Martínez-Álvarez C, Hedén P, Spalding KL, Vidal-Puig A, Medina-Gomez G, Arner P, Rydén M. Transforming Growth Factor-β3 Regulates Adipocyte Number in Subcutaneous White Adipose Tissue. Cell Rep 2018; 25:551-560.e5. [DOI: 10.1016/j.celrep.2018.09.069] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/31/2018] [Accepted: 09/21/2018] [Indexed: 11/15/2022] Open
|
49
|
Ben-Zvi D, Meoli L, Abidi WM, Nestoridi E, Panciotti C, Castillo E, Pizarro P, Shirley E, Gourash WF, Thompson CC, Munoz R, Clish CB, Anafi RC, Courcoulas AP, Stylopoulos N. Time-Dependent Molecular Responses Differ between Gastric Bypass and Dieting but Are Conserved Across Species. Cell Metab 2018; 28:310-323.e6. [PMID: 30043755 PMCID: PMC6628900 DOI: 10.1016/j.cmet.2018.06.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/19/2017] [Accepted: 06/04/2018] [Indexed: 12/22/2022]
Abstract
The effectiveness of Roux-en-Y gastric bypass (RYGB) against obesity and its comorbidities has generated excitement about developing new, less invasive treatments that use the same molecular mechanisms. Although controversial, RYGB-induced improvement of metabolic function may not depend entirely upon weight loss. To elucidate the differences between RYGB and dieting, we studied several individual organ molecular responses and generated an integrative, interorgan view of organismal physiology. We also compared murine and human molecular signatures. We show that, although dieting and RYGB can bring about the same degree of weight loss, post-RYGB physiology is very different. RYGB induces distinct, organ-specific adaptations in a temporal pattern that is characterized by energetically demanding processes, which may be coordinated by HIF1a activation and the systemic repression of growth hormone receptor signaling. Many of these responses are conserved in rodents and humans and may contribute to the remarkable ability of surgery to induce and sustain metabolic improvement.
Collapse
Affiliation(s)
- Danny Ben-Zvi
- Center for Basic and Translational Obesity Research, Division of Endocrinology, CLS16066, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Luca Meoli
- Center for Basic and Translational Obesity Research, Division of Endocrinology, CLS16066, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Wasif M Abidi
- Developmental Endoscopy Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Eirini Nestoridi
- Center for Basic and Translational Obesity Research, Division of Endocrinology, CLS16066, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Courtney Panciotti
- Center for Basic and Translational Obesity Research, Division of Endocrinology, CLS16066, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Erick Castillo
- Department of Digestive Surgery, School of Medicine, Pontificia Universidad Católica, Santiago 8331150, Chile
| | - Palmenia Pizarro
- Department of Digestive Surgery, School of Medicine, Pontificia Universidad Católica, Santiago 8331150, Chile
| | - Eleanor Shirley
- Division of Minimally Invasive and Metabolic Surgery, Magee-Womens Hospital, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - William F Gourash
- Division of Minimally Invasive and Metabolic Surgery, Magee-Womens Hospital, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Christopher C Thompson
- Developmental Endoscopy Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Rodrigo Munoz
- Department of Digestive Surgery, School of Medicine, Pontificia Universidad Católica, Santiago 8331150, Chile
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ron C Anafi
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anita P Courcoulas
- Division of Minimally Invasive and Metabolic Surgery, Magee-Womens Hospital, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Nicholas Stylopoulos
- Center for Basic and Translational Obesity Research, Division of Endocrinology, CLS16066, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
50
|
Stenkula KG, Erlanson-Albertsson C. Adipose cell size: importance in health and disease. Am J Physiol Regul Integr Comp Physiol 2018; 315:R284-R295. [DOI: 10.1152/ajpregu.00257.2017] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adipose tissue is necessary to harbor energy. To handle excess energy, adipose tissue expands by increasing adipocyte size (hypertrophy) and number (hyperplasia). Here, we have summarized the different experimental techniques used to study adipocyte cell size and describe adipocyte size in relation to insulin resistance, type 2 diabetes, and diet interventions. Hypertrophic adipocytes have an impaired cellular function, and inherent mechanisms restrict their expansion to protect against cell breakage and subsequent inflammation. Reduction of large fat cells by diet restriction, physical activity, or bariatric surgery therefore is necessary to improve cellular function and health. Small fat cells may also be dysfunctional and unable to expand. The distribution and function of the entire cell size range of fat cells, from small to very large fat cells, are an important but understudied aspect of adipose tissue biology. To prevent dysmetabolism, therapeutic strategies to expand small fat cells, recruit new fat cells, and reduce large fat cells are needed.
Collapse
Affiliation(s)
- Karin G. Stenkula
- Glucose Transport and Protein Trafficking, Biomedical Center, Lund University, Lund, Sweden
| | | |
Collapse
|