1
|
Loft A, Emont MP, Weinstock A, Divoux A, Ghosh A, Wagner A, Hertzel AV, Maniyadath B, Deplancke B, Liu B, Scheele C, Lumeng C, Ding C, Ma C, Wolfrum C, Strieder-Barboza C, Li C, Truong DD, Bernlohr DA, Stener-Victorin E, Kershaw EE, Yeger-Lotem E, Shamsi F, Hui HX, Camara H, Zhong J, Kalucka J, Ludwig JA, Semon JA, Jalkanen J, Whytock KL, Dumont KD, Sparks LM, Muir LA, Fang L, Massier L, Saraiva LR, Beyer MD, Jeschke MG, Mori MA, Boroni M, Walsh MJ, Patti ME, Lynes MD, Blüher M, Rydén M, Hamda N, Solimini NL, Mejhert N, Gao P, Gupta RK, Murphy R, Pirouzpanah S, Corvera S, Tang S, Das SK, Schmidt SF, Zhang T, Nelson TM, O'Sullivan TE, Efthymiou V, Wang W, Tong Y, Tseng YH, Mandrup S, Rosen ED. Towards a consensus atlas of human and mouse adipose tissue at single-cell resolution. Nat Metab 2025:10.1038/s42255-025-01296-9. [PMID: 40360756 DOI: 10.1038/s42255-025-01296-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/28/2025] [Indexed: 05/15/2025]
Abstract
Adipose tissue (AT) is a complex connective tissue with a high relative proportion of adipocytes, which are specialized cells with the ability to store lipids in large droplets. AT is found in multiple discrete depots throughout the body, where it serves as the primary repository for excess calories. In addition, AT has an important role in functions as diverse as insulation, immunity and regulation of metabolic homeostasis. The Human Cell Atlas Adipose Bionetwork was established to support the generation of single-cell atlases of human AT as well as the development of unified approaches and consensus for cell annotation. Here, we provide a first roadmap from this bionetwork, including our suggested cell annotations for humans and mice, with the aim of describing the state of the field and providing guidelines for the production, analysis, interpretation and presentation of AT single-cell data.
Collapse
Affiliation(s)
- Anne Loft
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark.
| | - Margo P Emont
- Section of Endocrinology, Diabetes and Metabolism, University of Chicago, Chicago, IL, USA.
| | - Ada Weinstock
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | - Adeline Divoux
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Adhideb Ghosh
- Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Allon Wagner
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Ann V Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, The University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Babukrishna Maniyadath
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Boxiang Liu
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular-Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Carey Lumeng
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Changhai Ding
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Chenkai Ma
- Human Health, Health and Biosecurity, CSIRO, Canberra, Australian Capital Territory, Australia
| | - Christian Wolfrum
- Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Clarissa Strieder-Barboza
- Department of Veterinary Sciences, Texas Tech University, Lubbock, TX, USA
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, USA
| | - Congru Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Danh D Truong
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, The University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | | | - Erin E Kershaw
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Esti Yeger-Lotem
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Farnaz Shamsi
- Department of Molecular Pathobiology, New York University, New York, NY, USA
- Departments of Cell Biology and Medicine, Grossman School of Medicine, New York University, New York, NY, USA
| | - Hannah X Hui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Henrique Camara
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Jiawei Zhong
- Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Joseph A Ludwig
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Julie A Semon
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Jutta Jalkanen
- Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Kyle D Dumont
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Lindsey A Muir
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Lucas Massier
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Luis R Saraiva
- Sidra Medicine, Doha, Qatar
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Marc D Beyer
- Immunogenomics and Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Platform for Single Cell Genomics and Epigenomics (PRECISE), German Center for Neurodegenerative Diseases (DZNE) and University of Bonn and West German Genome Center (WGGC), Bonn, Germany
| | - Marc G Jeschke
- Centre for Burn Research, Hamilton Health Sciences Centre, Department of Surgery and Department of Biochemistry, McMaster University, Hamilton, Ontario, Canada
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
- Obesity and Comorbidities Research Center (OCRC), Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Mariana Boroni
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Martin J Walsh
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary-Elizabeth Patti
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | | | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
- Department of Medicine - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, C2-94, Karolinska University Hospital, Stockholm, Sweden
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | | | - Nicole L Solimini
- Department of Medical Oncology, Sarcoma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Niklas Mejhert
- Department of Medicine (H7), Karolinska Institutet, C2-94, Karolinska University Hospital, Stockholm, Sweden
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Peng Gao
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rana K Gupta
- Department of Medicine, Division of Endocrinology, and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Saeed Pirouzpanah
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Silvia Corvera
- University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Su'an Tang
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Swapan K Das
- Department of Internal Medicine, Section on Endocrinology and Metabolism, Medical Center Boulevard, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Søren F Schmidt
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark
| | - Tao Zhang
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Theodore M Nelson
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Vissarion Efthymiou
- Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Wenjing Wang
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yihan Tong
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark.
| | - Evan D Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
2
|
Annunziata G, Verde L, Grillo ARA, Stallone T, Colao A, Muscogiuri G, Barrea L. Association among measurements obtained using portable ultrasonography with sex, body mass index, and age in a large sample of adult population. J Transl Med 2025; 23:236. [PMID: 40016819 PMCID: PMC11869708 DOI: 10.1186/s12967-025-06159-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/21/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND The assessment of the site-specific distribution of subcutaneous tissues (ST) represents an important addition to the body composition (BC) estimation. In this context, ultrasound stands out as a valuable method. However, the absence of reference values complicates both the interpretation of the obtained parameters and the monitoring of their changes during nutritional interventions or training programs. METHODS A total of 6,270 ultrasound images obtained with a technique using a non-diagnostic B-mode 2.5 MHz ultrasound probe (BodyMetrixTM, IntelaMetrix, Inc., Livermore, CA, USA) were obtained from adult men and women and analysed by grouping them according to sex, body mass index (BMI), and age. The thicknesses of total (tSAT), superficial (sSAT), and deep (dSAT) subcutaneous adipose tissue and muscle (MT), their ratios (sSAT/dSAT and MT/SAT), and muscle echogenicity (EG) were measured at the abdomen, thigh, chest, triceps, and hip sites. RESULTS Women exhibited greater tSAT, sSAT, dSAT, and EG (p < 0.001) and lower MT (p < 0.001), sSAT/dSAT (p = 0.008 for the abdomen and p < 0.001 for the thigh), and MT/SAT (p < 0.001) compared to men at both the abdomen and thigh sites. SAT-related parameters showed significant correlations with BMI, with thicknesses correlating positively and ratios negatively (p < 0.001). Muscle-related parameters correlated significantly with age, with MT and EG showing positive correlations and MT/SAT showing negative correlations (p < 0.001) in both sexes. Among SAT parameters, dSAT showed the strongest association with BMI in both men and women. CONCLUSIONS This study is the first to report the mean values of SAT-related quantitative and qualitative measurements (thickness and EG) in various body sites assessed through ultrasound and their associations with sex, BMI, and age in a large cohort. These findings could prove valuable in clinical practice for precise monitoring of nutritional and/or training interventions, as well as for a more comprehensive evaluation of nutritional status.
Collapse
Affiliation(s)
- Giuseppe Annunziata
- Facoltà di Scienze Umane, della Formazione e dello Sport, Università Telematica Pegaso, Via Porzio, Centro Direzionale, Isola F2, Naples, 80143, Italy
| | - Ludovica Verde
- Department of Public Health, University of Naples Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | | | - Tiziana Stallone
- Departmental Faculty of Medicine, UniCamillus-Saint Camillus International University of Health Sciences, Via Di Sant'Alessandro 8, Rome, 00131, Italy
| | - Annamaria Colao
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Endocrinologia, Diabetologia, Andrologia e Nutrizione, Unità di Endocrinologia, Diabetologia e Andrologia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Azienda Ospedaliera Universitaria Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", University Federico II, Naples, 80131, Italy
| | - Giovanna Muscogiuri
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Endocrinologia, Diabetologia, Andrologia e Nutrizione, Unità di Endocrinologia, Diabetologia e Andrologia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Azienda Ospedaliera Universitaria Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", University Federico II, Naples, 80131, Italy
| | - Luigi Barrea
- Dipartimento di Psicologia e Scienze della Salute, Università Telematica Pegaso, Centro Direzionale Isola F2, Via Porzio, Naples, 80143, Italy.
| |
Collapse
|
3
|
Yue Q, Cao Z, Li R, Wang Y, Rui S, Yin N, Liu L. Analysis of Physiological Oxygen Concentrations in Different Abdominal Fat Layers by Body Mass Index. Aesthetic Plast Surg 2025; 49:1145-1153. [PMID: 39466422 DOI: 10.1007/s00266-024-04479-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Physiological oxygen concentration in adipose tissue is closely linked to metabolic disorders such as chronic inflammation and insulin resistance. However, the nature of the variation in the oxygen levels of adipose tissue with body mass index (BMI) and depths of abdominal fat remain unclear. Therefore, this study aimed to elucidate the patterns of oxygen concentration in adipose tissue layers according to BMI. METHODS In this study, patients undergoing abdominal fat removal surgery were divided into the normal-weight (NW) or overweight-obese (OW) groups based on their BMI. Oxygen concentrations in abdominal superficial (sSAT) and deep subcutaneous adipose tissue (dSAT) were measured. The oxygen consumption rate, mean cell area, and capillary density in both tissue layers were compared between the two groups. Furthermore, the interaction between these three variables, BMI, and adipose tissue oxygen concentration, was analyzed using linear regression. RESULTS A total of 42 patients were recruited in this study and we observed that oxygen concentration in the sSAT was significantly lower than in the dSAT, irrespective of BMI. In terms of the oxygen concentration in the dSAT, OW's was significantly lower than that of NW's. Linear regression analysis revealed a significant correlation between dSAT oxygen concentration and BMI, mean adipocyte area, and vascular density. CONCLUSION Individuals who are obese have significantly lower oxygen levels in the deep abdominal adipose tissue, and this is influenced by BMI, adipocyte area, and capillary density. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Qiang Yue
- The Department of Perineal Plastic Surgery and Gender Reshaping of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Zilong Cao
- The Department of Perineal Plastic Surgery and Gender Reshaping of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Rui Li
- The Department of Perineal Plastic Surgery and Gender Reshaping of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Yunzhang Wang
- The Department of Perineal Plastic Surgery and Gender Reshaping of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Shu Rui
- The Department of Perineal Plastic Surgery and Gender Reshaping of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Ningbei Yin
- The Department of Cleft Lip and Palate of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China.
| | - Liqiang Liu
- The Department of Perineal Plastic Surgery and Gender Reshaping of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China.
| |
Collapse
|
4
|
Młynarska E, Czarnik W, Dzieża N, Jędraszak W, Majchrowicz G, Prusinowski F, Stabrawa M, Rysz J, Franczyk B. Type 2 Diabetes Mellitus: New Pathogenetic Mechanisms, Treatment and the Most Important Complications. Int J Mol Sci 2025; 26:1094. [PMID: 39940862 PMCID: PMC11817707 DOI: 10.3390/ijms26031094] [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: 12/20/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM), a prevalent chronic disease affecting over 400 million people globally, is driven by genetic and environmental factors. The pathogenesis involves insulin resistance and β-cell dysfunction, mediated by mechanisms such as the dedifferentiation of β-cells, mitochondrial dysfunction, and oxidative stress. Treatment should be based on non-pharmacological therapy. Strategies such as increased physical activity, dietary modifications, cognitive-behavioral therapy are important in maintaining normal glycemia. Advanced therapies, including SGLT2 inhibitors and GLP-1 receptor agonists, complement these treatments and offer solid glycemic control, weight control, and reduced cardiovascular risk. Complications of T2DM, such as diabetic kidney disease, retinopathy, and neuropathy, underscore the need for early diagnosis and comprehensive management to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Witold Czarnik
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Natasza Dzieża
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Weronika Jędraszak
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Gabriela Majchrowicz
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Filip Prusinowski
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Magdalena Stabrawa
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
5
|
de Zegher F, Díaz M, Ibáñez L. Ethnic variation in the relationship between ectopic fat and insulin resistance: Potential contributions of deep subcutaneous adipose tissue, rs1443512 in HOTAIR, and rs6712203 in COBLL1. Diabetes Obes Metab 2024; 26:6087-6088. [PMID: 39318051 DOI: 10.1111/dom.15980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024]
Affiliation(s)
- Francis de Zegher
- Leuven Research & Development, University of Leuven, Leuven, Belgium
| | - Marta Díaz
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Lourdes Ibáñez
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
6
|
Pakdee W, Laohawiriyakamol T, Tanutit P, Laohawiriyakamol S, Liabsuetrakul T. Association of body composition and survival in patients with locally advanced breast cancer: a historical cohort study. Acta Radiol 2024; 65:575-587. [PMID: 38591936 DOI: 10.1177/02841851241241528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
BACKGROUND Altered body composition has been attributed to major health problems globally, particularly in patients with cancer. To date, there have not been sufficient indices for body compositions in predicting the survival of locally advanced breast cancer (LABC). PURPOSE To assess the association between body composition and overall survival (OS) as well as disease-free survival (DFS) in patients with LABC. MATERIAL AND METHODS A retrospective study was conducted of patients with LABC diagnosed between 2010 and 2018. Body composition parameters, including skeletal muscle and adiposity parameters, were measured using computed tomography at the L3 vertebra using in-house software developed by MATLAB and freeware Python 3.6.13. The association between body composition and OS and DFS were analyzed using a log-rank test and multivariate Cox-proportional hazard regression. RESULTS Of 199 patients, 72 (36%) died during the follow-up period (range = 3.8-150.7 months). Median survival was 101 months. Low visceral-to-subcutaneous ratio ≤0.3 (adjusted hazard ratio [aHR] = 2.57, 95% confidence interval [CI] = 1.51-4.37; aHR = 2.46, 95% CI = 1.33-4.56), and high composite fat (aHR = 3.26, 95% CI = 1.69-6.29; aHR = 2.19, 95% CI = 1.11-4.3) were associated with lower OS and DFS. Positive lymph nodes ≥3, progesterone receptor negative, and total radiation dose >5000 cGy significantly decreased both OS and DFS. A history of previous treatment before body composition assessment and surgery had a protective effect on OS and DFS. No association of sarcopenia, body mass index, and adiposity areas with survival outcomes was observed. CONCLUSION Low visceral-to-subcutaneous ratio and high composite fat were independent prognostic factors for OS and DFS in patients with LABC. However, other body composition parameters showed no effect on survival.
Collapse
Affiliation(s)
- Wisitsak Pakdee
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | | | - Pramot Tanutit
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | | | - Tippawan Liabsuetrakul
- Department of Epidemiology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
7
|
Poltronieri TS, Pérsico RS, Viana LV. Body adipose tissue depots and treatment outcomes for women with breast cancer: A systematic review. Clin Nutr 2024; 43:1033-1042. [PMID: 38547637 DOI: 10.1016/j.clnu.2024.03.010] [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: 03/30/2023] [Revised: 02/14/2024] [Accepted: 03/16/2024] [Indexed: 05/04/2024]
Abstract
BACKGROUND & AIMS Excessive adipose tissue is associated with poorer prognosis in women with breast cancer (BC). However, several body adiposity depots, such as visceral (VAT), subcutaneous (SAT), intermuscular (IMAT), and gluteofemoral adipose tissues (GFAT) may have heterogeneous metabolic roles and health effects in these patients. This systematic review aims to evaluate the impact of different body adipose tissue depots, assessed via computed tomography (CT), on treatment outcomes for women with BC. We hypothesize that distinct body adipose tissue depots may be associated differently with outcomes in patients with BC. METHODS A comprehensive bibliographical search was conducted using PubMed, Embase, Cochrane Library, Scopus, and Web of Science databases (until January 2024). The methodological quality of the included studies was assessed using the Newcastle-Ottawa Scale. RESULTS The final sample comprised 23 retrospective studies (n = 12,462), with fourteen presenting good quality. A lack of standardization in CT body adipose tissue depots measurement and outcome presentation precluded quantitative analysis. Furthermore, most included studies had heterogeneous clinical characteristics. Survival and treatment response were the most prevalent outcomes. VAT (n = 19) and SAT (n = 17) were the most frequently evaluated depots and their increase was associated with worse outcomes, mainly in terms of survival. IMAT (n = 4) presented contradictory findings and a higher GFAT (n = 1) was associated with better outcomes. CONCLUSION This systematic review found an association between increased VAT and SAT with worse outcomes in patients with BC. However, due to the heterogeneity of the included studies, further research with homogeneous methodologies is necessary to better understand the impact of body adipose tissue depots on treatment outcomes. Such knowledge could lead to improved care for this patient population.
Collapse
Affiliation(s)
- Taiara Scopel Poltronieri
- Programa de Pós-Graduação em Ciências Médicas, Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Raquel Stocker Pérsico
- Programa de Pós-Graduação em Ciências Médicas, Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Luciana Verçoza Viana
- Programa de Pós-Graduação em Ciências Médicas, Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
8
|
Witczak-Sawczuk K, Ostrowska L, Cwalina U, Leszczyńska J, Jastrzębska-Mierzyńska M, Hładuński MK. Estimation of the Impact of Abdominal Adipose Tissue (Subcutaneous and Visceral) on the Occurrence of Carbohydrate and Lipid Metabolism Disorders in Patients with Obesity-A Pilot Study. Nutrients 2024; 16:1301. [PMID: 38732548 PMCID: PMC11085755 DOI: 10.3390/nu16091301] [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: 04/08/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Obesity represents a significant global public health concern. The excessive accumulation of abdominal adipose tissue is often implicated in the development of metabolic complications associated with obesity. Our study aimed to investigate the impact of particular deposits of abdominal adipose tissue on the occurrence of carbohydrate and lipid metabolism complications. We established cut-off points for visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT), and the VAT/SAT ratio at which selected metabolic complications of obesity-related diseases (disorders of carbohydrate and/or lipid metabolism) occur. We conducted an observational study involving 91 subjects with first- and second-degree obesity, accounting for gender differences. Anthropometric measurements were taken, body composition analysis (BIA) was conducted, and biochemical determinations were made. Our findings suggest that commonly used parameters for assessing early metabolic risk, such as BMI or waist circumference, may overlook the significant factor of body fat distribution, as well as gender differences. Both visceral and subcutaneous adipose tissue were found to be important in estimating metabolic risk. We identified the cut-off points in women in terms of their elevated fasting glucose levels and the presence of insulin resistance (HOMA-IR: homeostasis model assessment of insulin resistance) based on SAT, VAT, and the VAT/SAT ratio. In men, cut-off points were determined for the presence of insulin resistance (HOMA-IR) based on VAT and the VAT/SAT ratio. However, the results regarding lipid disorders were inconclusive, necessitating further investigation of a larger population.
Collapse
Affiliation(s)
- Katarzyna Witczak-Sawczuk
- Department of Dietetics and Clinical Nutrition, Medical University of Bialystok, ul. Mieszka I 4 B, 15-054 Bialystok, Poland; (K.W.-S.)
| | - Lucyna Ostrowska
- Department of Dietetics and Clinical Nutrition, Medical University of Bialystok, ul. Mieszka I 4 B, 15-054 Bialystok, Poland; (K.W.-S.)
| | - Urszula Cwalina
- Department of Biostatistics and Medical Informatics, Medical University of Bialystok, ul. Szpitalna 37, 15-295 Bialystok, Poland
| | - Joanna Leszczyńska
- Department of Dietetics and Clinical Nutrition, Medical University of Bialystok, ul. Mieszka I 4 B, 15-054 Bialystok, Poland; (K.W.-S.)
| | - Marta Jastrzębska-Mierzyńska
- Department of Dietetics and Clinical Nutrition, Medical University of Bialystok, ul. Mieszka I 4 B, 15-054 Bialystok, Poland; (K.W.-S.)
| | - Marcin Krzysztof Hładuński
- Independent Laboratory of Molecular Imaging, Medical University of Bialystok, ul. Zurawia 71A, 15-540 Bialystok, Poland
| |
Collapse
|
9
|
van den Munckhof ICL, Bahrar H, Schraa K, Brand T, Ter Horst R, van der Graaf M, Dekker HM, Stienstra R, de Graaf J, Joosten LAB, Netea MG, Riksen NP, Rutten JHW. Sex-specific association of visceral and subcutaneous adipose tissue volumes with systemic inflammation and innate immune cells in people living with obesity. Int J Obes (Lond) 2024; 48:523-532. [PMID: 38135702 DOI: 10.1038/s41366-023-01444-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 11/20/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND AND AIMS Obesity predisposes to metabolic and cardiovascular diseases. Adipose tissue inflammation and systemic inflammation contribute to these complications. There are strong sex differences in adipose tissue distribution and in systemic inflammation. Women have more subcutaneous adipose tissue (SAT) and less visceral adipose tissue (VAT) than men. We explored the sex differences in the association between the different adipose compartments and inflammatory markers that are important in cardiometabolic disease pathophysiology. METHODS Single-center observational cohort study with 302 individuals with a BMI ≥ 27 kg/m2. We were unable to acquire MRI data from seven individuals and from another 18 the MRI data were not usable, resulting in 277 people (155 men, 122 women), aged 55-81 years. INTERVENTION We performed the following measurements: abdominal magnetic resonance imaging to measure VAT, and SAT (deep and superficial) volumes; circulating leukocyte counts and cytokine production capacity of peripheral blood mononuclear cells (PBMCs), circulating cytokines, adipokines, and targeted proteomics; abdominal sSAT biopsies for histology and gene expression. RESULTS Only in women, (s)SAT volume was associated with circulating leukocytes, monocytes, and neutrophils. Circulating IL-6 and IL-18BP were associated with SAT volume in women and VAT in men. Several circulating proteins, including monocyte-colony-stimulating factor 1 and hepatocyte growth factor, are associated with sSAT in women and VAT in men. Only in women, SAT volume is associated with SAT expression of inflammatory proteins, including leptin, CD68, TNFα and IL-1α. CONCLUSION In women living with obesity, abdominal SAT volume, especially sSAT, is associated with circulating leukocytes and inflammatory proteins. In men, these parameters mainly show associations with VAT volume. This could be because only in women, sSAT volume is associated with sSAT expression of inflammatory proteins. These findings underscore that future research on adipose tissue in relation to cardiometabolic and cardiovascular disease should take sex differences into account.
Collapse
Affiliation(s)
| | - Harsh Bahrar
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kiki Schraa
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tessa Brand
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rob Ter Horst
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Helena M Dekker
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rinke Stienstra
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Jacqueline de Graaf
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115, Bonn, Germany
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
10
|
Polkinghorne MD, West HW, Antoniades C. Adipose Tissue in Cardiovascular Disease: From Basic Science to Clinical Translation. Annu Rev Physiol 2024; 86:175-198. [PMID: 37931169 DOI: 10.1146/annurev-physiol-042222-021346] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
The perception of adipose tissue as a metabolically quiescent tissue, primarily responsible for lipid storage and energy balance (with some endocrine, thermogenic, and insulation functions), has changed. It is now accepted that adipose tissue is a crucial regulator of metabolic health, maintaining bidirectional communication with other organs including the cardiovascular system. Additionally, adipose tissue depots are functionally and morphologically heterogeneous, acting not only as sources of bioactive molecules that regulate the physiological functioning of the vasculature and myocardium but also as biosensors of the paracrine and endocrine signals arising from these tissues. In this way, adipose tissue undergoes phenotypic switching in response to vascular and/or myocardial signals (proinflammatory, profibrotic, prolipolytic), a process that novel imaging technologies are able to visualize and quantify with implications for clinical prognosis. Furthermore, a range of therapeutic modalities have emerged targeting adipose tissue metabolism and altering its secretome, potentially benefiting those at risk of cardiovascular disease.
Collapse
Affiliation(s)
- Murray D Polkinghorne
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom;
- Acute Multidisciplinary Imaging and Interventional Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Henry W West
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom;
- Acute Multidisciplinary Imaging and Interventional Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Central Clinical School, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom;
- Acute Multidisciplinary Imaging and Interventional Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
11
|
de Paula Mancilha T, Massarani FA, Vieira F, Donangelo CM, Koury JC. Birth weight, skeletal maturity and dietary patterns are associated with body composition compartments differently in male and female physically active adolescents. Nutr Health 2023; 29:665-672. [PMID: 35491704 DOI: 10.1177/02601060221096514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Background: Adequate body composition is important for health and performance of physically active adolescents; however, some factors can influence body composition. Aim: The purpose of this study was to evaluate the association between body composition and birth weight, skeletal maturity and dietary patterns, in male (n = 124) and female (n = 107) physically active adolescents. Methods: Birth weight was obtained from health booklets. Dual-energy X-ray absorptiometry (DXA) was used to estimate fat mass (FM), fat-free mass (FFM), visceral adipose tissue (VAT) and abdominal adipose tissue (ASAT). Skeletal maturity was assessed by a hand and fist scanner using DXA. Food intake was assessed using a food frequency questionnaire and dietary patterns by factorial analysis. Results: Seventy one percent (male = 16%, female = 55%) of the participants were mature. Two dietary patterns (in natura/minimally processed and processed/ultra-processed) were identified. In males, FM (>15%, P = 0.043) and ASAT (>30%, P = 0.042) were higher in those with low-median compared to high-median birth weight. FFM (>24%) was higher in skeletally mature compared to those immatures (P = 0.001), and VAT (>28%) was higher in processed/ultra-processed compared to in natura/minimally processed dietary patterns (P = 0.015). In females, FFM (>9%) was higher in those with low-median compared to high-median birth weight (P = 0.019); and FFM (>16%), FM (>28%) and ASAT (>45%) were higher in skeletally mature compared to those immatures (P = 0.001). Conclusion: Our results may contribute to a better understanding of the complex association among body composition, birth weight, skeletal maturity and dietary patterns and in physically active adolescents, differently according to sex.
Collapse
Affiliation(s)
| | | | - Filomena Vieira
- Desporto e Saúde-Universidade de Lisboa Faculdade de Motricidade Humana, Lisboa, Portugal
| | | | - Josely Correa Koury
- Instituto de Nutrição-Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brasil
| |
Collapse
|
12
|
Kway YM, Thirumurugan K, Michael N, Tan KH, Godfrey KM, Gluckman P, Chong YS, Venkataraman K, Khoo EYH, Khoo CM, Leow MKS, Tai ES, Chan JK, Chan SY, Eriksson JG, Fortier MV, Lee YS, Velan SS, Feng M, Sadananthan SA. A fully convolutional neural network for comprehensive compartmentalization of abdominal adipose tissue compartments in MRI. Comput Biol Med 2023; 167:107608. [PMID: 37897959 DOI: 10.1016/j.compbiomed.2023.107608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/18/2023] [Accepted: 10/17/2023] [Indexed: 10/30/2023]
Abstract
BACKGROUND Existing literature has highlighted structural, physiological, and pathological disparities among abdominal adipose tissue (AAT) sub-depots. Accurate separation and quantification of these sub-depots are crucial for advancing our understanding of obesity and its comorbidities. However, the absence of clear boundaries between the sub-depots in medical imaging data has challenged their separation, particularly for internal adipose tissue (IAT) sub-depots. To date, the quantification of AAT sub-depots remains challenging, marked by a time-consuming, costly, and complex process. PURPOSE To implement and evaluate a convolutional neural network to enable granular assessment of AAT by compartmentalization of subcutaneous adipose tissue (SAT) into superficial subcutaneous (SSAT) and deep subcutaneous (DSAT) adipose tissue, and IAT into intraperitoneal (IPAT), retroperitoneal (RPAT), and paraspinal (PSAT) adipose tissue. MATERIAL AND METHODS MRI datasets were retrospectively collected from Singapore Preconception Study for Long-Term Maternal and Child Outcomes (S-PRESTO: 389 women aged 31.4 ± 3.9 years) and Singapore Adult Metabolism Study (SAMS: 50 men aged 28.7 ± 5.7 years). For all datasets, ground truth segmentation masks were created through manual segmentation. A Res-Net based 3D-UNet was trained and evaluated via 5-fold cross-validation on S-PRESTO data (N = 300). The model's final performance was assessed on a hold-out (N = 89) and an external test set (N = 50, SAMS). RESULTS The proposed method enabled reliable segmentation of individual AAT sub-depots in 3D MRI volumes with high mean Dice similarity scores of 98.3%, 97.2%, 96.5%, 96.3%, and 95.9% for SSAT, DSAT, IPAT, RPAT, and PSAT respectively. CONCLUSION Convolutional neural networks can accurately sub-divide abdominal SAT into SSAT and DSAT, and abdominal IAT into IPAT, RPAT, and PSAT with high accuracy. The presented method has the potential to significantly contribute to advancements in the field of obesity imaging and precision medicine.
Collapse
Affiliation(s)
- Yeshe M Kway
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kashthuri Thirumurugan
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore
| | - Navin Michael
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore
| | - Kok Hian Tan
- Duke-National University of Singapore Graduate Medical School, Singapore; Department of Maternal Fetal Medicine, KK Women's and Children's Hospital, Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Centre & NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Peter Gluckman
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kavita Venkataraman
- Saw Swee Hock School of Public Health, National University of Singapore, National University Health System, Singapore
| | - Eric Yin Hao Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chin Meng Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Medicine, National University Health System, Singapore
| | - Melvin Khee-Shing Leow
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore; Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Endocrinology, Division of Medicine, Tan Tock Seng Hospital (TTSH), Singapore
| | - E Shyong Tai
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Endocrinology, University Medicine Cluster, National University Health System, Singapore
| | - Jerry Ky Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore; Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Johan G Eriksson
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Folkhälsan Research Center, Helsinki, Finland
| | - Marielle V Fortier
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore; Department of Diagnostic and Interventional Imaging, KK Women's and Children's Hospital, Singapore
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Paediatric Endocrinology, Department of Paediatrics, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore
| | - S Sendhil Velan
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore
| | - Mengling Feng
- Saw Swee Hock School of Public Health, National University of Singapore, National University Health System, Singapore; Institute of Data Science, National University of Singapore, Singapore
| | - Suresh Anand Sadananthan
- Singapore Institute for Clinical Sciences, Agency for Science Technology, and Research, Singapore.
| |
Collapse
|
13
|
Guo Y, Hu Z, Chen J, Zhang J, Fan Z, Qu Q, Miao Y. Feasibility of adipose-derived therapies for hair regeneration: Insights based on signaling interplay and clinical overview. J Am Acad Dermatol 2023; 89:784-794. [PMID: 34883154 DOI: 10.1016/j.jaad.2021.11.058] [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: 02/17/2021] [Revised: 09/13/2021] [Accepted: 11/30/2021] [Indexed: 11/22/2022]
Abstract
Dermal white adipose tissue (dWAT) is a dynamic component of the skin and closely interacts with the hair follicle. Interestingly, dWAT envelops the hair follicle during anagen and undergoes fluctuations in volume throughout the hair cycle. dWAT-derived extracellular vesicles can significantly regulate the hair cycle, and this provides a theoretical basis for utilizing adipose tissue as a feasible clinical strategy to treat hair loss. However, the amount and depth of the available literature are far from enough to fully elucidate the prominent role of dWAT in modulating the hair growth cycle. This review starts by investigating the hair cycle-coupled dWAT remodeling and the reciprocal signaling interplay underneath. Then, it summarizes the current literature and assesses the advantages and limitations of clinical research utilizing adipose-derived therapies for hair regeneration.
Collapse
Affiliation(s)
- Yilong Guo
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jian Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jiarui Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhexiang Fan
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Qian Qu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Yong Miao
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
14
|
Blaak EE, Goossens GH. Metabolic phenotyping in people living with obesity: Implications for dietary prevention. Rev Endocr Metab Disord 2023; 24:825-838. [PMID: 37581871 PMCID: PMC10492670 DOI: 10.1007/s11154-023-09830-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2023] [Indexed: 08/16/2023]
Abstract
Given the increasing number of people living with obesity and related chronic metabolic disease, precision nutrition approaches are required to increase the effectiveness of prevention strategies. This review addresses these approaches in different metabolic phenotypes (metabotypes) in obesity. Although obesity is typically associated with an increased cardiometabolic disease risk, some people with obesity are relatively protected against the detrimental effects of excess adiposity on cardiometabolic health, also referred to as 'metabolically healthy obesity' (MHO). Underlying mechanisms, the extent to which MHO is a transient state as well as lifestyle strategies to counteract the transition from MHO to metabolically unhealthy obesity (MUO) are discussed. Based on the limited resources that are available for dietary lifestyle interventions, it may be reasonable to prioritize interventions for people with MUO, since targeting high-risk patients for specific nutritional, lifestyle or weight-loss strategies may enhance the cost-effectiveness of these interventions. Additionally, the concept of tissue insulin resistant (IR) metabotypes is discussed, representing distinct etiologies towards type 2 diabetes (T2D) as well as cardiovascular disease (CVD). Recent evidence indicates that these tissue IR metabotypes, already present in individuals with obesity with a normal glucose homeostasis, respond differentially to diet. Modulation of dietary macronutrient composition according to these metabotypes may considerably improve cardiometabolic health benefits. Thus, nutritional or lifestyle intervention may improve cardiometabolic health, even with only minor or no weight loss, which stresses the importance of focusing on a healthy lifestyle and not on weight loss only. Targeting different metabotypes towards T2D and cardiometabolic diseases may lead to more effective lifestyle prevention and treatment strategies. Age and sex-related differences in tissue metabotypes and related microbial composition and functionality (fermentation), as important drivers and/or mediators of dietary intervention response, have to be taken into account. For the implementation of these approaches, more prospective trials are required to provide the knowledge base for precision nutrition in the prevention of chronic metabolic diseases.
Collapse
Affiliation(s)
- Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
15
|
Carli F, Sabatini S, Gaggini M, Sironi AM, Bedogni G, Gastaldelli A. Fatty Liver Index (FLI) Identifies Not Only Individuals with Liver Steatosis but Also at High Cardiometabolic Risk. Int J Mol Sci 2023; 24:14651. [PMID: 37834099 PMCID: PMC10572624 DOI: 10.3390/ijms241914651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
A fatty liver index (FLI) greater than sixty (FLI ≥ 60) is an established score for metabolic dysfunction-associated steatotic liver disease (MASLD), which carries a high risk for diabetes and cardiovascular disease, while a FLI ≤ 20 rules out the presence of steatosis. Thus, we investigated whether FLI was associated with cardiometabolic risk factors, i.e., visceral (VAT), subcutaneous (SC), epicardial (EPI), extrapericardial (PERI), and total cardiac (CARD-AT) adipose tissue, hepatic fat ((by magnetic resonance imaging, MRI, and spectroscopy, MRS), and insulin resistance (IR, HOMA-IR and OGIS-index), and components of metabolic syndrome. All individuals with FLI ≥ 60 had MASLD, while none with FLI ≤ 20 had steatosis (by MRS). Subjects with FLI ≥ 60 had a higher BMI and visceral and cardiac fat (VAT > 1.7 kg, CARD-AT > 0.2 kg). FLI was positively associated with increased cardiac and visceral fat and components of metabolic syndrome. FLI, VAT, and CARD-AT were all associated with IR, increased blood pressure, cholesterol, and reduced HDL. For FLI ≥ 60, the cut-off values for fat depots and laboratory measures were estimated. In conclusion, FLI ≥ 60 identified not only subjects with steatosis but also those with IR, abdominal and cardiac fat accumulation, increased blood pressure, and hyperlipidemia, i.e., those at higher risk of cardiometabolic diseases. Targeted reduction of FLI components would help reduce cardiometabolic risk.
Collapse
Affiliation(s)
- Fabrizia Carli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council (CNR), Via Giuseppe Moruzzi, 1, 56124 Pisa, Italy; (F.C.); (S.S.); (M.G.); (A.M.S.)
| | - Silvia Sabatini
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council (CNR), Via Giuseppe Moruzzi, 1, 56124 Pisa, Italy; (F.C.); (S.S.); (M.G.); (A.M.S.)
| | - Melania Gaggini
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council (CNR), Via Giuseppe Moruzzi, 1, 56124 Pisa, Italy; (F.C.); (S.S.); (M.G.); (A.M.S.)
| | - Anna Maria Sironi
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council (CNR), Via Giuseppe Moruzzi, 1, 56124 Pisa, Italy; (F.C.); (S.S.); (M.G.); (A.M.S.)
| | - Giorgio Bedogni
- Department of Medical and Surgical Sciences, University of Bologna, Via Zamboni, 33, 40126 Bologna, Italy
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council (CNR), Via Giuseppe Moruzzi, 1, 56124 Pisa, Italy; (F.C.); (S.S.); (M.G.); (A.M.S.)
| |
Collapse
|
16
|
Cesaro A, De Michele G, Fimiani F, Acerbo V, Scherillo G, Signore G, Rotolo FP, Scialla F, Raucci G, Panico D, Gragnano F, Moscarella E, Scudiero O, Mennitti C, Calabrò P. Visceral adipose tissue and residual cardiovascular risk: a pathological link and new therapeutic options. Front Cardiovasc Med 2023; 10:1187735. [PMID: 37576108 PMCID: PMC10421666 DOI: 10.3389/fcvm.2023.1187735] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Obesity is a heterogeneous disease that affects almost one-third of the global population. A clear association has been established between obesity and cardiovascular disease (CVD). However, CVD risk is known to be related more to the local distribution of fat than to total body fat. Visceral adipose tissue (VAT) in particular has a high impact on CVD risk. This manuscript reviews the role of VAT in residual CV risk and the available therapeutic strategies for decreasing residual CV risk related to VAT accumulation. Among the many pathways involved in residual CV risk, obesity and particularly VAT accumulation play a major role by generating low-grade systemic inflammation, which in turn has a high prognostic impact on all-cause mortality and myocardial infarction. In recent years, many therapeutic approaches have been developed to reduce body weight. Orlistat was shown to reduce both weight and VAT but has low tolerability and many drug-drug interactions. Naltrexone-bupropion combination lowers body weight but has frequent side effects and is contraindicated in patients with uncontrolled hypertension. Liraglutide and semaglutide, glucagon-like peptide 1 (GLP-1) agonists, are the latest drugs approved for the treatment of obesity, and both have been shown to induce significant body weight loss. Liraglutide, semaglutide and other GLP-1 agonists also showed a positive effect on CV outcomes in diabetic patients. In addition, liraglutide showed to specifically reduce VAT and inflammatory biomarkers in obese patients without diabetes. GLP-1 agonists are promising compounds to limit inflammation in human visceral adipocytes.
Collapse
Affiliation(s)
- Arturo Cesaro
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Gianantonio De Michele
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Fabio Fimiani
- Unit of Inherited and Rare Cardiovascular Diseases, A.O.R.N. Dei Colli “V. Monaldi”, Naples, Italy
| | - Vincenzo Acerbo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Gianmaria Scherillo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Giovanni Signore
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Francesco Paolo Rotolo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Francesco Scialla
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Giuseppe Raucci
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Domenico Panico
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Elisabetta Moscarella
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Olga Scudiero
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
- Ceinge Biotecnologie Avanzate Franco Salvatore S. C. a R. L., Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Cristina Mennitti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| |
Collapse
|
17
|
Baptista LS, Silva KR, Jobeili L, Guillot L, Sigaudo-Roussel D. Unraveling White Adipose Tissue Heterogeneity and Obesity by Adipose Stem/Stromal Cell Biology and 3D Culture Models. Cells 2023; 12:1583. [PMID: 37371053 PMCID: PMC10296800 DOI: 10.3390/cells12121583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
The immune and endocrine dysfunctions of white adipose tissue are a hallmark of metabolic disorders such as obesity and type 2 diabetes. In humans, white adipose tissue comprises distinct depots broadly distributed under the skin (hypodermis) and as internal depots (visceral). Depot-specific ASCs could account for visceral and subcutaneous adipose tissue properties, by regulating adipogenesis and immunomodulation. More importantly, visceral and subcutaneous depots account for distinct contributions to obesity and its metabolic comorbidities. Recently, distinct ASCs subpopulations were also described in subcutaneous adipose tissue. Interestingly, the superficial layer closer to the dermis shows hyperplastic and angiogenic capacities, whereas the deep layer is considered as having inflammatory properties similar to visceral. The aim of this focus review is to bring the light of recent discoveries into white adipose tissue heterogeneity together with the biology of distinct ASCs subpopulations and to explore adipose tissue 3D models revealing their advantages, disadvantages, and contributions to elucidate the role of ASCs in obesity development. Recent advances in adipose tissue organoids opened an avenue of possibilities to recreate the main cellular and molecular events of obesity leading to a deep understanding of this inflammatory disease besides contributing to drug discovery. Furthermore, 3D organ-on-a-chip will add reproducibility to these adipose tissue models contributing to their translation to the pharmaceutical industry.
Collapse
Affiliation(s)
- Leandra S. Baptista
- Numpex-bio, Campus UFRJ Duque de Caxias Prof Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rio de Janeiro 25240005, Brazil
| | - Karina R. Silva
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550900, Brazil;
- Teaching and Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940070, Brazil
| | - Lara Jobeili
- Laboratory of Tissue Biology and Therapeutic Engineering, University of Lyon, Claude Bernard University Lyon 1, CNRS, LBTI UMR 5305, 69367 Lyon, France; (L.J.); (L.G.); (D.S.-R.)
| | - Lucile Guillot
- Laboratory of Tissue Biology and Therapeutic Engineering, University of Lyon, Claude Bernard University Lyon 1, CNRS, LBTI UMR 5305, 69367 Lyon, France; (L.J.); (L.G.); (D.S.-R.)
- Urgo Research Innovation and Development, 21300 Chenôve, France
| | - Dominique Sigaudo-Roussel
- Laboratory of Tissue Biology and Therapeutic Engineering, University of Lyon, Claude Bernard University Lyon 1, CNRS, LBTI UMR 5305, 69367 Lyon, France; (L.J.); (L.G.); (D.S.-R.)
| |
Collapse
|
18
|
Montazeri M, Zarkesh M, Zadeh-Vakili A, Khalili D, Movahedi M, Khalaj A. Association of physical activity with increased PI3K and Akt mRNA levels in adipose tissues of obese and non-obese adults. Sci Rep 2023; 13:9291. [PMID: 37286617 DOI: 10.1038/s41598-023-36365-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 06/02/2023] [Indexed: 06/09/2023] Open
Abstract
Phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathway regulates glucose and lipid metabolism. We examined the association of PI3K and Akt expression in visceral (VAT) and subcutaneous adipose tissue (SAT) with daily physical activity (PA) in non-diabetic obese and non-obese adults. In this cross-sectional study, we included 105 obese (BMI ≥ 30 kg/m2) and 71 non-obese (BMI < 30 kg/m2) subjects (aged/ ≥ 18 years). PA was measured using a valid and reliable International Physical Activity Questionnaire(IPAQ)-long-form, and the metabolic equivalent of task(MET) was calculated. Real-time PCR was performed to analyze the mRNA relative expression. VAT PI3K expression had a lower level in obese compared to non-obese (P = 0.015), while its expression was higher in active individuals than inactive ones (P = 0.029). SAT PI3K expression was increased in active individuals compared to inactive ones (P = 0.031). There was a rise in VAT Akt expression in the actives compared to the inactive participants (P = 0.037) and in non-obese/active compared to non-obese/inactive individuals (P = 0.026). Obese individuals had a decreased expression level of SAT Akt compared to non-obsesses (P = 0.005). VAT PI3K was directly and significantly associated with PA in obsesses (β = 1.457, P = 0.015). Positive association between PI3K and PA suggests beneficial effects of PA for obese individuals that can be partly described by PI3K/Akt pathway acceleration in adipose tissue.
Collapse
Affiliation(s)
- Marzieh Montazeri
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Zarkesh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, PO Box: 19395-4763, Tehran, Iran.
| | - Azita Zadeh-Vakili
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, PO Box: 19395-4763, Tehran, Iran.
| | - Davood Khalili
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Monireh Movahedi
- Department of Biochemistry, Faculty of Biological Science, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Alireza Khalaj
- Tehran Obesity Treatment Center, Department of Surgery, Shahed University, Tehran, Iran
| |
Collapse
|
19
|
Ishiuchi N, Nakashima A, Maeda S, Miura Y, Miyasako K, Sasaki K, Uchiki T, Sasaki A, Nagamatsu S, Nakao N, Nagao M, Masaki T. Comparison of therapeutic effects of mesenchymal stem cells derived from superficial and deep subcutaneous adipose tissues. Stem Cell Res Ther 2023; 14:121. [PMID: 37143086 PMCID: PMC10161523 DOI: 10.1186/s13287-023-03350-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 04/19/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Fibrosis is a common histological feature in the process from chronic organ injury to organ failure. Chronic tissue injury causes inflammatory cell infiltration into the injured tissue. The persistence of this inflammatory cell infiltration leads to fibrosis and organ failure. Adipose-derived mesenchymal stem cells (ASCs) have received much attention as a regenerative therapeutic tool to prevent progression from organ injury to failure. Subcutaneous abdominal adipose tissue is divided into superficial and deep layers by a superficial fascia. Adipose tissue easily collected by liposuction is usually obtained from a deep layer, so ASCs derived from a deep layer are generally used for regenerative medicine. However, no research has been conducted to investigate differences in the therapeutic effects of ASCs from the superficial and deep layers (Sup-ASCs and Deep-ASCs, respectively). Therefore, we compared the therapeutic potencies of Sup-ASCs and Deep-ASCs. METHODS ASCs were isolated from superficial and deep subcutaneous abdominal adipose tissues collected from patients who underwent breast reconstruction. We first compared cell characteristics, such as morphology, cell proliferation, cell surface markers, adipogenic and osteogenic differentiation, cell senescence markers, and expression of coagulation and anticoagulant factors between Sup-ASCs and Deep-ASCs. Furthermore, we compared their ability to promote polarization of M2 macrophages and to inhibit transforming growth factor (TGF)-β/Smad signaling using THP-1 cells and TGF-β1 stimulated HK-2 cells incubated with conditioned media from Sup-ASCs or Deep-ASCs. In in vivo experiments, after renal ischemia-reperfusion injury (IRI) procedure, Sup-ASCs or Deep-ASCs were injected through the abdominal aorta. At 21 days post-injection, the rats were sacrificed and their left kidneys were collected to evaluate fibrosis. Finally, we performed RNA-sequencing analysis of Sup-ASCs and Deep-ASCs. RESULTS Sup-ASCs had greater proliferation and adipogenic differentiation compared with Deep-ASCs, whereas both ASC types had similar morphology, cell surface markers, senescence markers, and expression of coagulation and anticoagulant factors. Conditioned media from Sup-ASCs and Deep-ASCs equally promoted polarization of M2 macrophages and suppressed TGF-β/Smad signaling. Moreover, administration of Sup-ASCs and Deep-ASCs equally ameliorated renal fibrosis induced by IRI in rats. RNA-sequencing analysis revealed no significant difference in the expression of genes involved in anti-inflammatory and anti-fibrotic effects between Sup-ASCs and Deep-ASCs. CONCLUSIONS These results indicate that both Sup-ASCs and Deep-ASCs can be used effectively and safely as an intravascular ASC therapy for organ injury.
Collapse
Affiliation(s)
- Naoki Ishiuchi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
- Center for Cause of Death Investigation Research, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- Department of Forensic Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Satoshi Maeda
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- TWOCELLS Company, Limited, 16-35 Hijiyama-honmachi, Minami-ku, Hiroshima, 732-0816, Japan
| | - Yoshie Miura
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- TWOCELLS Company, Limited, 16-35 Hijiyama-honmachi, Minami-ku, Hiroshima, 732-0816, Japan
| | - Kisho Miyasako
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kensuke Sasaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Toshio Uchiki
- Department of Plastic and Reconstructive Surgery, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Ayano Sasaki
- Department of Plastic and Reconstructive Surgery, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shogo Nagamatsu
- Department of Plastic and Reconstructive Surgery, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Naoki Nakao
- Department of Forensic Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Masataka Nagao
- Center for Cause of Death Investigation Research, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- Department of Forensic Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| |
Collapse
|
20
|
de Ritter R, Sep SJS, van Greevenbroek MMJ, Kusters YHAM, Vos RC, Bots ML, Kooi ME, Dagnelie PC, Eussen SJPM, Schram MT, Koster A, Brouwers MCG, van der Sangen NMR, Peters SAE, van der Kallen CJH, Stehouwer CDA. Sex differences in body composition in people with prediabetes and type 2 diabetes as compared with people with normal glucose metabolism: the Maastricht Study. Diabetologia 2023; 66:861-872. [PMID: 36805778 PMCID: PMC10036428 DOI: 10.1007/s00125-023-05880-0] [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: 05/27/2022] [Accepted: 11/22/2022] [Indexed: 02/23/2023]
Abstract
AIMS/HYPOTHESIS Obesity is a major risk factor for type 2 diabetes. However, body composition differs between women and men. In this study we investigate the association between diabetes status and body composition and whether this association is moderated by sex. METHODS In a population-based cohort study (n=7639; age 40-75 years, 50% women, 25% type 2 diabetes), we estimated the sex-specific associations, and differences therein, of prediabetes (i.e. impaired fasting glucose and/or impaired glucose tolerance) and type 2 diabetes (reference: normal glucose metabolism [NGM]) with dual-energy x-ray absorptiometry (DEXA)- and MRI-derived measures of body composition and with hip circumference. Sex differences were analysed using adjusted regression models with interaction terms of sex-by-diabetes status. RESULTS Compared with their NGM counterparts, both women and men with prediabetes and type 2 diabetes had more fat and lean mass and a greater hip circumference. The differences in subcutaneous adipose tissue, hip circumference and total and peripheral lean mass between type 2 diabetes and NGM were greater in women than men (women minus men [W-M] mean difference [95% CI]: 15.0 cm2 [1.5, 28.5], 3.2 cm [2.2, 4.1], 690 g [8, 1372] and 443 g [142, 744], respectively). The difference in visceral adipose tissue between type 2 diabetes and NGM was greater in men than women (W-M mean difference [95% CI]: -14.8 cm2 [-26.4, -3.1]). There was no sex difference in the percentage of liver fat between type 2 diabetes and NGM. The differences in measures of body composition between prediabetes and NGM were generally in the same direction, but were not significantly different between women and men. CONCLUSIONS/INTERPRETATION This study indicates that there are sex differences in body composition associated with type 2 diabetes. The pathophysiological significance of these sex-associated differences requires further study.
Collapse
Affiliation(s)
- Rianneke de Ritter
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands.
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands.
| | - Simone J S Sep
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
- Adelante, Center of Expertise in Rehabilitation and Audiology, Hoensbroek, the Netherlands
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Yvo H A M Kusters
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Rimke C Vos
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Leiden University Medical Center, Department of Public Health and Primary Care/LUMC-Campus, The Hague, the Netherlands
| | - Michiel L Bots
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - M Eline Kooi
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Pieter C Dagnelie
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Simone J P M Eussen
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
- Department of Epidemiology, Maastricht University, Maastricht, the Netherlands
- CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
| | - Miranda T Schram
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
- Heart and Vascular Center, Maastricht University Medical Center+, Maastricht, the Netherlands
- MHeNs School for Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Annemarie Koster
- CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
- Department of Social Medicine, Maastricht University, Maastricht, the Netherlands
| | - Martijn C G Brouwers
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | | | - Sanne A E Peters
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- The George Institute for Global Health, Imperial College London, London, UK
| | - Carla J H van der Kallen
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
21
|
Wu H, Li Q, Zhang K, Zhao J. The association between serum copper and obesity and all-cause mortality: the NHANES 2011-2016. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:31395-31407. [PMID: 36449235 DOI: 10.1007/s11356-022-24432-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
Excessive serum copper has multiple effects on human health, while the association between copper and obesity remains unclear. The objective of this study is to examine the associations of serum copper concentrations with obesity and adiposity measures, including body fat composition and distribution among adults in the USA. This analysis utilized data from the National Health and Nutrition Examination Survey (NHANES) (2011-2016). A total of 7285 adults aged 18 years or older who had serum copper measures were included in this cross-sectional study. Multi-linear regression and logistic regression were used to estimate the independent risky effect of copper on fat deposition and all-cause mortality. Moreover, these associations were analyzed in stratification analysis by gender, age, and physical activity (PA). Generally, we found that participants who were females, non-Hispanic Black, or with inactive PA tended to have a higher serum level of copper. In addition, we observed positive associations between serum copper and adiposity measurements. Furthermore, a serum copper level higher than 133.9 μg/dL was a risk factor for all-cause mortality, which doubled the odds ratio of all-cause mortality compared to the normal serum copper level. Serum copper was positively associated with fat deposition of whole body and regional parts, and all-cause mortality. Furthermore, the effects of copper on fat distribution were also significant and could be modified by age, gender, and PA.
Collapse
Affiliation(s)
- Hongrong Wu
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| | - Qingqi Li
- Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Kaifang Zhang
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| | - Jianfeng Zhao
- Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, People's Republic of China.
| |
Collapse
|
22
|
Shen W, Middleton MS, Cunha GM, Delgado TI, Wolfson T, Gamst A, Fowler KJ, Alazraki A, Trout AT, Ohliger MA, Shah SN, Bashir MR, Kleiner DE, Loomba R, Neuschwander-Tetri BA, Sanyal AJ, Zhou J, Sirlin CB, Lavine JE. Changes in abdominal adipose tissue depots assessed by MRI correlate with hepatic histologic improvement in non-alcoholic steatohepatitis. J Hepatol 2023; 78:238-246. [PMID: 36368598 PMCID: PMC9852022 DOI: 10.1016/j.jhep.2022.10.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND & AIMS Non-alcoholic steatohepatitis (NASH) is prevalent in adults with obesity and can progress to cirrhosis. In a secondary analysis of prospectively acquired data from the multicenter, randomized, placebo-controlled FLINT trial, we investigated the relationship between reduction in adipose tissue compartment volumes and hepatic histologic improvement. METHODS Adult participants in the FLINT trial with paired liver biopsies and abdominal MRI exams at baseline and end-of-treatment (72 weeks) were included (n = 76). Adipose tissue compartment volumes were obtained using MRI. RESULTS Treatment and placebo groups did not differ in baseline adipose tissue volumes, or in change in adipose tissue volumes longitudinally (p = 0.107 to 0.745). Deep subcutaneous adipose tissue (dSAT) and visceral adipose tissue volume reductions were associated with histologic improvement in NASH (i.e., NAS [non-alcoholic fatty liver disease activity score] reductions of ≥2 points, at least 1 point from lobular inflammation and hepatocellular ballooning, and no worsening of fibrosis) (p = 0.031, and 0.030, respectively). In a stepwise logistic regression procedure, which included demographics, treatment group, baseline histology, baseline and changes in adipose tissue volumes, MRI hepatic proton density fat fraction (PDFF), and serum aminotransferases as potential predictors, reductions in dSAT and PDFF were associated with histologic improvement in NASH (regression coefficient = -2.001 and -0.083, p = 0.044 and 0.033, respectively). CONCLUSIONS In adults with NASH in the FLINT trial, those with greater longitudinal reductions in dSAT and potentially visceral adipose tissue volumes showed greater hepatic histologic improvements, independent of reductions in hepatic PDFF. CLINICAL TRIAL NUMBER NCT01265498. IMPACT AND IMPLICATIONS Although central obesity has been identified as a risk factor for obesity-related disorders including insulin resistance and cardiovascular disease, the role of central obesity in non-alcoholic steatohepatitis (NASH) warrants further clarification. Our results highlight that a reduction in central obesity, specifically deep subcutaneous adipose tissue and visceral adipose tissue, may be related to histologic improvement in NASH. The findings from this analysis should increase awareness of the importance of lifestyle intervention in NASH for clinical researchers and clinicians. Future studies and clinical practice may design interventions that assess the reduction of deep subcutaneous adipose tissue and visceral adipose tissue as outcome measures, rather than simply weight reduction.
Collapse
Affiliation(s)
- Wei Shen
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA;; Institute of Human Nutrition, College of Physicians & Surgeons, Columbia University Irving Medical Center; NY, USA;; Columbia Magnetic Resonance Research Center (CMRRC), Columbia University, USA.
| | - Michael S Middleton
- Liver Imaging Group, Department of Radiology, UCSD School of Medicine, San Diego, CA, USA
| | | | - Timoteo I Delgado
- Liver Imaging Group, Department of Radiology, UCSD School of Medicine, San Diego, CA, USA
| | - Tanya Wolfson
- Computational and Applied Statistics Laboratory (CASL), San Diego Supercomputer Center at UCSD, San Diego, CA, USA
| | - Anthony Gamst
- Computational and Applied Statistics Laboratory (CASL), San Diego Supercomputer Center at UCSD, San Diego, CA, USA;; Department of Mathematics, UCSD, San Diego, CA, USA
| | - Kathryn J Fowler
- Liver Imaging Group, Department of Radiology, UCSD School of Medicine, San Diego, CA, USA
| | - Adina Alazraki
- Emory University School of Medicine, Department of Radiology and Imaging Sciences and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Andrew T Trout
- Department of Radiology, Cincinnati Children's Hospital Medical Center and Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael A Ohliger
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Shetal N Shah
- Section of Abdominal Imaging and Nuclear Medicine Department, Imaging Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mustafa R Bashir
- Department of Radiology, Duke University Medical Center, Durham, North Carolina, USA;; Center for Advanced Magnetic Resonance Development, (CAMRD), Department of Radiology, Duke University Medical Center, Durham, NC, USA;; Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - David E Kleiner
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology, Department of Medicine, University of California-San Diego, La Jolla, CA, USA
| | | | | | - Jane Zhou
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Claude B Sirlin
- Liver Imaging Group, Department of Radiology, UCSD School of Medicine, San Diego, CA, USA
| | - Joel E Lavine
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA;; Institute of Human Nutrition, College of Physicians & Surgeons, Columbia University Irving Medical Center; NY, USA
| |
Collapse
|
23
|
Trinh L, Stenkula KG, Olsson LE, Svensson J, Peterson P, Bennet L, Månsson S. Favorable fatty acid composition in adipose tissue in healthy Iraqi- compared to Swedish-born men - a pilot study using MRI assessment. Adipocyte 2022; 11:153-163. [PMID: 35291924 PMCID: PMC8928862 DOI: 10.1080/21623945.2022.2042963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Middle Eastern immigrants are at high-risk for insulin resistance. Fatty acid composition (FAC) plays an important role in the development of insulin resistance but has not been investigated in people of Middle Eastern ancestry. Here, the aim was to assess the FAC in visceral and subcutaneous adipose tissue (VAT and SAT) in healthy Iraqi- and Swedish-born men using a magnetic resonance imaging (MRI) method.This case-control study included 23 Iraqi- and 15 Swedish-born middle-aged men, without cardiometabolic disease. Using multi-echo MRI of the abdomen, the fractions of saturated, monounsaturated, and polyunsaturated fatty acids (fSFA, fMUFA, and fPUFA) were estimated in VAT and SAT. SAT was further analyzed in deep and superficial compartments (dSAT and sSAT). In all depots, fPUFA was significantly higher and fSFA significantly lower in Iraqi men, independently of age and BMI. In both Iraqi- and Swedish-born men, higher fPUFA and lower fMUFA were found in sSAT vs. dSAT. Among Iraqi men only, higher fPUFA and lower fMUFA were found in SAT vs. VAT.Iraqi-born men presented a more favorable abdominal FAC compared to Swedish-born men. This MRI method also revealed different FACs in different abdominal depots. Our results may reflect a beneficial FAC in Middle Eastern immigrants.
Collapse
Affiliation(s)
- Lena Trinh
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Karin G Stenkula
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Lars E Olsson
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Malmö, Sweden
- Hematology, Oncology and Radiation Physics, Skåne University Hospital, Malmö, Sweden
| | - Jonas Svensson
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Malmö, Sweden
- Medical Imaging and Physiology, Skåne University Hospital, Lund, Sweden
| | - Pernilla Peterson
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Malmö, Sweden
- Medical Imaging and Physiology, Skåne University Hospital, Lund, Sweden
| | - Louise Bennet
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Clinical Research and Trial Centre, Lund University Hospital, Lund, Sweden
| | - Sven Månsson
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Malmö, Sweden
- Hematology, Oncology and Radiation Physics, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
24
|
Nikolic M, Novakovic J, Ramenskaya G, Kokorekin V, Jeremic N, Jakovljevic V. Cooling down with Entresto. Can sacubitril/valsartan combination enhance browning more than coldness? Diabetol Metab Syndr 2022; 14:175. [PMID: 36419097 PMCID: PMC9686067 DOI: 10.1186/s13098-022-00944-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND It is a growing importance to induce a new treatment approach to encourage weight loss but also to improve maintenance of lost weight. It has been shown that promotion of brown adipose tissue (BAT) function or acquisition of BAT characteristics in white adipose tissue (terms referred as "browning") can be protective against obesity. MAIN TEXT Amongst numerous established environmental influences on BAT activity, cold exposure is the best interested technique due to its not only effects on of BAT depots in proliferation process but also de novo differentiation of precursor cells via β-adrenergic receptor activation. A novel combination drug, sacubitril/valsartan, has been shown to be more efficient in reducing cardiovascular events and heart failure readmission compared to conventional therapy. Also, this combination of drugs increases the postprandial lipid oxidation contributing to energy expenditure, promotes lipolysis in adipocytes and reduces body weight. To date, there is no research examining potential of combined sacubitril/valsartan use to promote browning or mechanisms in the basis of this thermogenic process. CONCLUSION Due to the pronounced effects of cold and sacubitril/valsartan treatment on function and metabolism of BAT, the primary goal of further research should focused on investigation of the synergistic effects of the sacubitril/valsartan treatment at low temperature environmental conditions.
Collapse
Affiliation(s)
- Marina Nikolic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Jovana Novakovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | | | - Nevena Jeremic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
- First Moscow State Medical University IM Sechenov, Moscow, Russia.
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Human Pathology, First Moscow State Medical University IM Sechenov, Moscow, Russia
| |
Collapse
|
25
|
Li Y, Wang H, Chen H, Liao Y, Gou S, Yan Q, Zhuang Z, Li H, Wang J, Suo Y, Lan T, Liu Y, Zhao Y, Zou Q, Nie T, Hui X, Lai L, Wu D, Fan N. Generation of a genetically modified pig model with CREBRF R457Q variant. FASEB J 2022; 36:e22611. [PMID: 36250915 DOI: 10.1096/fj.202201117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/03/2022] [Indexed: 11/11/2022]
Abstract
Obesity is among the strongest risk factors for type 2 diabetes (T2D). The CREBRF missense allele rs373863828 (p. Arg457Gln, p. R457Q) is associated with increased body mass index but reduced risk of T2D in people of Pacific ancestry. To investigate the functional consequences of the CREBRF variant, we introduced the corresponding human mutation R457Q into the porcine genome. The CREBRFR457Q pigs displayed dramatically increased fat deposition, which was mainly distributed in subcutaneous adipose tissue other than visceral adipose tissue. The CREBRFR457Q variant promoted preadipocyte differentiation. The increased differentiation capacity of precursor adipocytes conferred pigs the unique histological phenotype that adipocytes had a smaller size but a greater number in subcutaneous adipose tissue (SAT) of CREBRFR457Q variant pigs. In addition, in SAT of CREBRFR457Q pigs, the contents of the peroxidative metabolites 4-hydroxy-nonenal and malondialdehyde were significantly decreased, while the activity of antioxidant enzymes, such as glutathione peroxidase, superoxide dismutase, and catalase, was increased, which was in accordance with the declined level of the reactive oxygen species (ROS) in CREBRFR457Q pigs. Together, these data supported a causal role of the CREBRFR457Q variant in the pathogenesis of obesity, partly via adipocyte hyperplasia, and further suggested that reduced oxidative stress in adipose tissue may mediate the relative metabolic protection afforded by this variant despite the related obesity.
Collapse
Affiliation(s)
- Yingying Li
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Hai Wang
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Huangyao Chen
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yuan Liao
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Shixue Gou
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Quanmei Yan
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhenpeng Zhuang
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Hao Li
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Jiaowei Wang
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yangyang Suo
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Ting Lan
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yang Liu
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yu Zhao
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Qingjian Zou
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Tao Nie
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaoyan Hui
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong SAR
| | - Liangxue Lai
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Donghai Wu
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Nana Fan
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
26
|
Sun W, Zhang T, Hu S, Tang Q, Long X, Yang X, Gun S, Chen L. Chromatin accessibility landscape of stromal subpopulations reveals distinct metabolic and inflammatory features of porcine subcutaneous and visceral adipose tissue. PeerJ 2022; 10:e13250. [PMID: 35646489 PMCID: PMC9138157 DOI: 10.7717/peerj.13250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/21/2022] [Indexed: 01/13/2023] Open
Abstract
Background Fat accumulation in visceral adipose tissue (VAT) confers increased risk for metabolic disorders of obesity, whereas accumulation of subcutaneous adipose tissue (SAT) is associated with lower risk and may be protective. Previous studies have shed light on the gene expression profile differences between SAT and VAT; however, the chromatin accessibility landscape differences and how the cis-regulatory elements govern gene expression changes between SAT and VAT are unknown. Methods Pig were used to characterize the differences in chromatin accessibility between the two adipose depots-derived stromal vascular fractions (SVFs) using DNase-sequencing (DNase-seq). Using integrated data from DNase-seq, H3K27ac ChIP-sequencing (ChIP-seq), and RNA-sequencing (RNA-seq), we investigated how the regulatory locus complexity regulated gene expression changes between SAT and VAT and the possible impact that these changes may have on the different biological functions of these two adipose depots. Results SVFs form SAT and VAT (S-SVF and V-SVF) have differential chromatin accessibility landscapes. The differential DNase I hypersensitive site (DHS)-associated genes, which indicate dynamic chromatin accessibility, were mainly involved in metabolic processes and inflammatory responses. Additionally, the Krüppel-like factor family of transcription factors were enriched in the differential DHSs. Furthermore, the chromatin accessibility data were highly associated with differential gene expression as indicated using H3K27ac ChIP-seq and RNA-seq data, supporting the validity of the differential gene expression determined using DNase-seq. Moreover, by combining epigenetic and transcriptomic data, we identified two candidate genes, NR1D1 and CRYM, could be crucial to regulate distinct metabolic and inflammatory characteristics between SAT and VAT. Together, these results uncovered differences in the transcription regulatory network and enriched the mechanistic understanding of the different biological functions between SAT and VAT.
Collapse
Affiliation(s)
- Wenyang Sun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China,Key Laboratory of Pig Industry Sciences (Ministry of Agriculture), Chongqing Academy of Animal Science, Chongqing, China
| | - Tinghuan Zhang
- Key Laboratory of Pig Industry Sciences (Ministry of Agriculture), Chongqing Academy of Animal Science, Chongqing, China
| | - Silu Hu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qianzi Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xi Long
- Key Laboratory of Pig Industry Sciences (Ministry of Agriculture), Chongqing Academy of Animal Science, Chongqing, China
| | - Xu Yang
- College of Nursing, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Lei Chen
- Key Laboratory of Pig Industry Sciences (Ministry of Agriculture), Chongqing Academy of Animal Science, Chongqing, China
| |
Collapse
|
27
|
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
|
28
|
Li M, Zheng Q, Miller JD, Zuo P, Yuan X, Feng J, Liu C, Bao S, Lou Q. Aerobic training reduces pancreatic fat content and improves β-cell function: A randomized controlled trial using IDEAL-IQ magnetic resonance imaging. Diabetes Metab Res Rev 2022; 38:e3516. [PMID: 34963031 DOI: 10.1002/dmrr.3516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/14/2021] [Accepted: 12/08/2021] [Indexed: 11/09/2022]
Abstract
AIMS To explore the effects of six months of moderate-intensity aerobic exercise on pancreatic fat content and its impact on β-cell function. MATERIALS AND METHODS A total of 106 patients with type 2 diabetes mellitus were randomized to either a moderate-intensity aerobic training group (three times a week, including 5 min warm-up, 50 min aerobic dancing, and 5 min relaxation, n = 53) or control group (n = 53) with 6-month intervention. The primary endpoint was change in pancreatic fat content. An intention-to-treat analysis was conducted. RESULTS Eighty-six patients completed the study with 43 patients in the aerobic training group. The average age, HbA1c, and pancreatic fat content for all participants (106 patients) were 66.39 ± 5.59 years, 7.05 ± 1.24%, and 10.35 ± 9.20%, respectively. Nearly half (49.06%) of patients were males. Subjects in the aerobic training group saw a significant reduction in pancreatic fat content when compared to controls (p = 0.001). In logistic regression models containing age, diabetes duration, change in BMI, smoking/drinking status, changes in lipid indices, and other abdominal fat content, only reduction in pancreatic fat content (p < 0.05) was an independent protective factor for β-cell function and HbA1c. CONCLUSIONS Six months of moderate-intensity aerobic training significantly reduced the pancreatic fat content. The reduction of pancreatic fat content was an independent protective factor for β-cell function and HbA1c.
Collapse
Affiliation(s)
- Min Li
- Department of Endocrinology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- Nursing College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qidong Zheng
- Department of Internal Medicine, Yuhuan Second People's Hospital, Yuhuan, China
| | - Joshua D Miller
- Division of Endocrinology and Metabolism, Department of Medicine, Renaissance School of Medicine at Stony Brook University, New York, USA
| | - Panpan Zuo
- School of Nursing, Taizhou Polytehnic College, Taizhou, China
| | - Xiaodan Yuan
- Department of Healthy Education, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jitao Feng
- Department of Radiology, Nanjing Pukou Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Chao Liu
- Research Center of Endocrine and Metabolic Diseases, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shan Bao
- Department of Gynaecology and Obstetrics, Hainan General Hospital, Haikou, China
| | - Qingqing Lou
- Department of Endocrinology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
29
|
Kahn DE, Bergman BC. Keeping It Local in Metabolic Disease: Adipose Tissue Paracrine Signaling and Insulin Resistance. Diabetes 2022; 71:599-609. [PMID: 35316835 PMCID: PMC8965661 DOI: 10.2337/dbi21-0020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 01/03/2022] [Indexed: 01/04/2023]
Abstract
Alterations in adipose tissue composition and function are associated with obesity and contribute to the development of type 2 diabetes. While the significance of this relationship has been cemented, our understanding of the multifaceted role of adipose tissue in metabolic heath and disease continues to evolve and expand. Heterogenous populations of cells that make up adipose tissue throughout the body generate diverse secretomes containing a mosaic of bioactive compounds with vast structural and signaling capabilities. While there are many reports highlighting the important role of adipose tissue endocrine signaling in insulin resistance and type 2 diabetes, the direct, local, paracrine effect of adipose tissue has received less attention. Recent studies have begun to underscore the importance of considering anatomically discrete adipose depots for their specific impact on local microenvironments and metabolic function in neighboring tissues as well as regulation of whole-body physiology. This article highlights the important role of adipose tissue paracrine signaling on metabolic function and insulin sensitivity in nearby tissues and organs, specifically focusing on visceral, pancreatic, subcutaneous, intermuscular, and perivascular adipose tissue depots.
Collapse
Affiliation(s)
- Darcy E. Kahn
- University of Colorado Anschutz Medical Campus, Aurora, CO
| | | |
Collapse
|
30
|
Liu X, Liu L, Zhao J, Wang H, Li Y. Mechanotransduction regulates inflammation responses of epicardial adipocytes in cardiovascular diseases. Front Endocrinol (Lausanne) 2022; 13:1080383. [PMID: 36589802 PMCID: PMC9800500 DOI: 10.3389/fendo.2022.1080383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue is a crucial regulator in maintaining cardiovascular homeostasis by secreting various bioactive products to mediate the physiological function of the cardiovascular system. Accumulating evidence shows that adipose tissue disorders contribute to several kinds of cardiovascular disease (CVD). Furthermore, the adipose tissue would present various biological effects depending on its tissue localization and metabolic statuses, deciding the individual cardiometabolic risk. Crosstalk between adipose and myocardial tissue is involved in the pathophysiological process of arrhythmogenic right ventricular cardiomyopathy (ARVC), cardiac fibrosis, heart failure, and myocardial infarction/atherosclerosis. The abnormal distribution of adipose tissue in the heart might yield direct and/or indirect effects on cardiac function. Moreover, mechanical transduction is critical for adipocytes in differentiation, proliferation, functional maturity, and homeostasis maintenance. Therefore, understanding the features of mechanotransduction pathways in the cellular ontogeny of adipose tissue is vital for underlining the development of adipocytes involved in cardiovascular disorders, which would preliminarily contribute positive implications on a novel therapeutic invention for cardiovascular diseases. In this review, we aim to clarify the role of mechanical stress in cardiac adipocyte homeostasis and its interplay with maintaining cardiac function.
Collapse
Affiliation(s)
- Xiaoliang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junfei Zhao
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| | - Hua Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| |
Collapse
|
31
|
Singh P, Covassin N, Marlatt K, Gadde KM, Heymsfield SB. Obesity, Body Composition, and Sex Hormones: Implications for Cardiovascular Risk. Compr Physiol 2021; 12:2949-2993. [PMID: 34964120 PMCID: PMC10068688 DOI: 10.1002/cphy.c210014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Cardiovascular disease (CVD) continues to be the leading cause of death in adults, highlighting the need to develop novel strategies to mitigate cardiovascular risk. The advancing obesity epidemic is now threatening the gains in CVD risk reduction brought about by contemporary pharmaceutical and surgical interventions. There are sex differences in the development and outcomes of CVD; premenopausal women have significantly lower CVD risk than men of the same age, but women lose this advantage as they transition to menopause, an observation suggesting potential role of sex hormones in determining CVD risk. Clear differences in obesity and regional fat distribution among men and women also exist. While men have relatively high fat in the abdominal area, women tend to distribute a larger proportion of their fat in the lower body. Considering that regional body fat distribution is an important CVD risk factor, differences in how men and women store their body fat may partly contribute to sex-based alterations in CVD risk as well. This article presents findings related to the role of obesity and sex hormones in determining CVD risk. Evidence for the role of sex hormones in determining body composition in men and women is also presented. Lastly, the clinical potential for using sex hormones to alter body composition and reduce CVD risk is outlined. © 2022 American Physiological Society. Compr Physiol 12:1-45, 2022.
Collapse
Affiliation(s)
- Prachi Singh
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | | | - Kara Marlatt
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Kishore M Gadde
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Steven B Heymsfield
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| |
Collapse
|
32
|
Gomez-Perez SL, Zhang Y, Mourtzakis M, Tussing-Humphreys L, Ridlon J, Gaskins HR, Mutlu E. Comparison between handheld ultrasound and regional and whole-body dual energy x-ray absorptiometry (DXA) for body fat assessment. Clin Nutr ESPEN 2021; 46:386-393. [PMID: 34857225 DOI: 10.1016/j.clnesp.2021.08.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 01/06/2023]
Abstract
OBJECTIVES To determine the extent of agreement between a handheld ultrasound (US) attached to an android tablet and the reference method dual energy x-ray absorptiometry (DXA) for the measurement of adiposity. METHODS A whole-body DXA scan and abdominal adipose tissue thickness measurements using a handheld US were obtained from 104 adults (63 females, 41 males). Body fat percent (BF%), total fat mass (kg), and trunk fat mass (kg) were obtained from DXA. Subcutaneous adipose tissue (SAT), superficial subcutaneous adipose tissue (SSAT), and deep subcutaneous adipose tissue (DSAT) thickness were obtained from US. Sex-specific total fat mass, trunk fat mass, and BF% estimates by US were compared with DXA. Spearman's correlations and Bland-Altman plots were used to assess agreement between the methods. RESULTS US SAT correlated strongly with total fat mass for both females (rs = 0.74) and males (rs = 0.87) as did trunk fat mass (females, rs = 0.81; males, rs = 0.83); as did SSAT and DSAT (females: rs = 0.65 and rs = 0.66; males: rs = 0.63 and rs = 0.85, respectively, all p-values < 0.0001). Bland-Altman plots demonstrated strong agreement for total and trunk fat mass for both males and females. For BF%, acceptable limits of agreement were observed for males but not for females, substantial proportional bias as indicated by a negative slope was noted for BF% using SAT (r = -0.298, p = 0.0177). CONCLUSION The handheld US and technique to analyze abdominal adipose tissue thickness showed strong agreement with DXA results and generated highly comparable estimates for total and trunk fat mass for both sexes.
Collapse
Affiliation(s)
| | - Yanyu Zhang
- Rush University Medical Center, Rush Bioinformatics and Biostatistics Core, Chicago, IL, USA
| | - Marina Mourtzakis
- University of Waterloo, Department of Kinesiology, Waterloo, Ontario, Canada
| | - Lisa Tussing-Humphreys
- University of Illinois at Chicago, Department of Kinesiology and Nutrition, UIC Cancer Center, Chicago, IL, USA
| | - Jason Ridlon
- University of Illinois Urbana-Champaign, Department of Animal Sciences, Urbana-Champaign, IL, USA; Cancer Center at Illinois, Urbana-Champaign, IL, USA
| | - H Rex Gaskins
- University of Illinois Urbana-Champaign, Department of Animal Sciences, Urbana-Champaign, IL, USA; Cancer Center at Illinois, Urbana-Champaign, IL, USA
| | - Ece Mutlu
- Rush University Medical Center, Department of Medicine, Chicago, IL, USA
| |
Collapse
|
33
|
Diaz-Canestro C, Xu A. Impact of Different Adipose Depots on Cardiovascular Disease. J Cardiovasc Pharmacol 2021; 78:S30-S39. [PMID: 34840259 DOI: 10.1097/fjc.0000000000001131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/05/2021] [Indexed: 12/13/2022]
Abstract
ABSTRACT Adipose tissue (AT)-derived factors contribute to the regulation of cardiovascular homeostasis, thereby playing an important role in cardiovascular health and disease. In obesity, AT expands and becomes dysfunctional, shifting its secretory profile toward a proinflammatory state associated with deleterious effects on the cardiovascular system. AT in distinct locations (ie, adipose depots) differs in crucial phenotypic variables, including inflammatory and secretory profile, cellular composition, lipolytic activity, and gene expression. Such heterogeneity among different adipose depots may explain contrasting cardiometabolic risks associated with different obesity phenotypes. In this respect, central obesity, defined as the accumulation of AT in the abdominal region, leads to higher risk of cardiometabolic alterations compared with the accumulation of AT in the gluteofemoral region (ie, peripheral obesity). The aim of this review was to provide an updated summary of clinical and experimental evidence supporting the differential roles of different adipose depots in cardiovascular disease and to discuss the molecular basis underlying the differences of adipose depots in the regulation of cardiovascular function.
Collapse
Affiliation(s)
- Candela Diaz-Canestro
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
- Department of Medicine, the University of Hong Kong, Hong Kong, China; and
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
- Department of Medicine, the University of Hong Kong, Hong Kong, China; and
- Department of Pharmacology and Pharmacy, the University of Hong Kong, Hong Kong, China
| |
Collapse
|
34
|
Gowri S M, Antonisamy B, Geethanjali FS, Thomas N, Jebasingh F, Paul TV, Karpe F, Osmond C, Fall CHD, Vasan SK. Distinct opposing associations of upper and lower body fat depots with metabolic and cardiovascular disease risk markers. Int J Obes (Lond) 2021; 45:2490-2498. [PMID: 34331002 DOI: 10.1038/s41366-021-00923-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/03/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND To examine the associations of total and regional adiposity with metabolic and cardiovascular disease (CVD) risk markers. METHODS This cross-sectional study included 1080 (53.8% men, aged 39-44 years) individuals from South India. Anthropometry (height, weight, waist and hip circumference), body composition assessment using dual-energy X-ray absorptiometry (DXA), blood pressure (BP), and plasma glucose, insulin and lipids were measured. Regression analysis was used to examine associations of standardized fat measurements with type 2 diabetes (T2D), insulin resistance (IR), hypertension and hypertriglyceridemia and continuous measurements of BP, glucose, insulin, HOMA-IR and lipids. Contour plots were constructed to visualize the differential effect of upper and lower fat depots. RESULTS DXA-measured fat depots were positively associated with metabolic and CVD risk markers. After adjusting for fat mass index, upper body fat remained positively, while lower body fat was negatively associated with risk markers. A one standard deviation (SD) increase in android fat showed higher odds ratios (ORs) for T2D (6.59; 95% CI 3.17, 13.70), IR (4.68; 95% CI 2.31, 9.50), hypertension (2.57; 95% CI 1.56, 4.25) and hypertriglyceridemia (6.39; 95% CI 3.46, 11.90) in men. A 1 SD increase in leg fat showed a protective effect with ORs for T2D (0.42; 95% CI 0.24, 0.74), IR (0.31; 95% CI 0.17, 0.57) and hypertriglyceridemia (0.61; 95% CI 0.38, 0.98). The magnitude of the effect was greater with DXA-measured fat compared with anthropometry. CONCLUSION At any level of total body fat, upper and lower body fat depots demonstrate opposite risk associations with metabolic and CVD risk markers in Asian Indians.
Collapse
Affiliation(s)
| | | | | | - Nihal Thomas
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, India
| | - Felix Jebasingh
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, India
| | - Thomas V Paul
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, India
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Clive Osmond
- MRC Environmental Epidemiology Unit, Southampton General Hospital, University of Southampton, Southampton, UK
| | - Caroline H D Fall
- MRC Environmental Epidemiology Unit, Southampton General Hospital, University of Southampton, Southampton, UK
| | - Senthil K Vasan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK.
- MRC Environmental Epidemiology Unit, Southampton General Hospital, University of Southampton, Southampton, UK.
| |
Collapse
|
35
|
Kastberg SE, Lund HS, de Lucia-Rolfe E, Kaduka LU, Boit MK, Corpeleijn E, Friis H, Bernard S, Paquette M, Baas A, Rasmussen JJ, Christensen DL. Hepatic steatosis is associated with anthropometry, cardio-metabolic disease risk, sex, age and urbanisation, but not with ethnicity in adult Kenyans. Trop Med Int Health 2021; 27:49-57. [PMID: 34704339 PMCID: PMC7614816 DOI: 10.1111/tmi.13696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE We aimed to determine the associations of non-alcoholic fatty liver disease (NAFLD) with cardio-metabolic risk factors for diabetes in adult Kenyans. METHODS A cross-sectional study was undertaken among rural and urban Kenyans of different ethnic origin. Ultrasonography scanning (USS) methods were used for the assessment of hepatic fat accumulation for NAFLD assessment and abdominal fat distribution, and simple anthropometry measurements were performed. All participants underwent a 2-h oral glucose tolerance test, and biochemical, haemodynamic and lifestyle data were obtained. Multivariate logistic regression analyses were used to assess sex, age, residency and ethnic differences in the association between NAFLD and various metabolic parameters. RESULTS In total, 743 individuals (59.1% women) with a mean age of 38.0 (range 18-68) years participated in the study. Overall, 118 individuals (15.9%) had NAFLD, of whom 94.1% had mild steatosis. Age >40 years was significantly associated with having NAFLD compared with <30 years of age with no difference found in NAFLD between ethnic groups (Luo, Kamba, Maasai). All body composition and clinical measurements were associated with NAFLD (p < 0.045 for OR). CONCLUSION Finding lower odds for NAFLD in men was unexpected, as was the lack of differences in NAFLD among the ethnic groups, while higher odds for NAFLD with increasing age and in urban vs. rural populations was expected. Especially the sex-specific results warrant further studies in black African populations on biology of body composition for having NAFLD, and whether this translates into insulin resistance and higher risk of diabetes and consequently cardiovascular disease in black African women.
Collapse
Affiliation(s)
- Sophie E Kastberg
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Helene S Lund
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Emanuella de Lucia-Rolfe
- NIHR Cambridge Biomedical Research Centre-Diet, Anthropometry and Physical Activity Group, MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Lydia U Kaduka
- Centre for Public Health Research, KEMRI, Nairobi, Kenya
| | - Michael K Boit
- Department of Physical, Exercise and Sport Science, Kenyatta University, Nairobi, Kenya
| | - Eva Corpeleijn
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Henrik Friis
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Sophie Bernard
- Lipids, Nutrition and Cardiovascular Prevention Clinic of the Montreal Clinical Research Institute, Montreal, Canada
| | - Martine Paquette
- Lipids, Nutrition and Cardiovascular Prevention Clinic of the Montreal Clinical Research Institute, Montreal, Canada
| | - Alexis Baas
- Lipids, Nutrition and Cardiovascular Prevention Clinic of the Montreal Clinical Research Institute, Montreal, Canada.,Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Montreal, Canada
| | - Jon J Rasmussen
- Department of Endocrinology, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Dirk L Christensen
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Paris MT, Bell KE, Avrutin E, Mourtzakis M. Associations between skeletal muscle echo intensity and thickness in relation to glucose homeostasis in healthy and glucose impaired older males. Exp Gerontol 2021; 154:111547. [PMID: 34506901 DOI: 10.1016/j.exger.2021.111547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/18/2021] [Accepted: 08/30/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Aging-related changes in muscle composition and mass may predispose older adults to developing insulin resistance. Ultrasound echo intensity and thickness are surrogates of muscle composition and mass, however, their associations with glucose homeostasis are not well established. We examined how muscle echo intensity and thickness correlate with markers of glucose homeostasis in older (≥65 years) males with normal (n = 22) or impaired (n = 10) glucose control. METHODS Echo intensity was measured for the biceps brachii, rectus abdominis, and rectus femoris. Muscle thickness was evaluated for the biceps brachii + brachioradialis, rectus abdominis, and rectus femoris + vastus intermedius. Glucose homeostasis was evaluated using a 2-h oral glucose tolerance test. RESULTS In older males with normal glucose homeostasis, higher echo intensity of the rectus abdominis and rectus femoris was moderately (r = 0.36 to 0.59) associated with 2-h glucose. On the contrary, higher muscle echo intensity of the rectus abdominis, biceps brachii, and rectus femoris was moderately-to-strongly (r = -0.36 to -0.79) associated with indices of better glucose homeostasis in the impaired group. Rectus abdominis muscle thickness was moderately associated (r = 0.36) with better glucose tolerance in the normal glucose homeostasis; however, in the glucose impaired group, muscle thickness was associated with (r = 0.37 to 0.73) with poorer glucose homeostasis. CONCLUSIONS Muscle echo intensity displays divergent associations with glucose homeostasis in older males with normal compared to impaired glucose control. Larger muscle thickness was associated with poorer glucose homeostasis in the glucose impaired group, but rectus abdominis muscle thickness was correlated with better homeostasis in healthy older males.
Collapse
Affiliation(s)
- Michael T Paris
- Department of Kinesiology, University of Waterloo, Waterloo, ON, Canada
| | - Kirsten E Bell
- Department of Kinesiology, University of Waterloo, Waterloo, ON, Canada
| | - Egor Avrutin
- Department of Kinesiology, University of Waterloo, Waterloo, ON, Canada
| | - Marina Mourtzakis
- Department of Kinesiology, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
37
|
Kway YM, Thirumurugan K, Tint MT, Michael N, Shek LPC, Yap FKP, Tan KH, Godfrey KM, Chong YS, Fortier MV, Marx UC, Eriksson JG, Lee YS, Velan SS, Feng M, Sadananthan SA. Automated Segmentation of Visceral, Deep Subcutaneous, and Superficial Subcutaneous Adipose Tissue Volumes in MRI of Neonates and Young Children. Radiol Artif Intell 2021; 3:e200304. [PMID: 34617030 DOI: 10.1148/ryai.2021200304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 06/01/2021] [Accepted: 07/12/2021] [Indexed: 11/11/2022]
Abstract
Purpose To develop and evaluate an automated segmentation method for accurate quantification of abdominal adipose tissue (AAT) depots (superficial subcutaneous adipose tissue [SSAT], deep subcutaneous adipose tissue [DSAT], and visceral adipose tissue [VAT]) in neonates and young children. Materials and Methods This was a secondary analysis of prospectively collected data, which used abdominal MRI data from Growing Up in Singapore Towards healthy Outcomes, or GUSTO, a longitudinal mother-offspring cohort, to train and evaluate a convolutional neural network for volumetric AAT segmentation. The data comprised imaging volumes of 333 neonates obtained at early infancy (age ≤2 weeks, 180 male neonates) and 755 children aged either 4.5 years (n = 316, 150 male children) or 6 years (n = 439, 219 male children). The network was trained on images of 761 randomly selected volumes (neonates and children combined) and evaluated on 100 neonatal volumes and 227 child volumes by using 10-fold validation. Automated segmentations were compared with expert-generated manual segmentation. Segmentation performance was assessed using Dice scores. Results When the model was tested on the test datasets across the 10 folds, the model had strong agreement with the ground truth for all testing sets, with mean Dice similarity scores for SSAT, DSAT, and VAT, respectively, of 0.960, 0.909, and 0.872 in neonates and 0.944, 0.851, and 0.960 in children. The model generalized well to different body sizes and ages and to all abdominal levels. Conclusion The proposed segmentation approach provided accurate automated volumetric assessment of AAT compartments on MR images of neonates and children.Keywords Pediatrics, Deep Learning, Convolutional Neural Networks, Water-Fat MRI, Image Segmentation, Deep and Superficial Subcutaneous Adipose Tissue, Visceral Adipose TissueClinical trial registration no. NCT01174875 Supplemental material is available for this article. © RSNA, 2021.
Collapse
Affiliation(s)
- Yeshe Manuel Kway
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Kashthuri Thirumurugan
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Mya Thway Tint
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Navin Michael
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Lynette Pei-Chi Shek
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Fabian Kok Peng Yap
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Kok Hian Tan
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Keith M Godfrey
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Marielle Valerie Fortier
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Ute C Marx
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Johan G Eriksson
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - S Sendhil Velan
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Mengling Feng
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| | - Suresh Anand Sadananthan
- Singapore Institute for Clinical Sciences (Y.M.K., K.T., M.T.T., N.M., L.P.C.S., Y.S.C., M.V.F., J.G.E., Y.S.L., S.S.V., S.A.S.) and Institute of Bioengineering and Bioimaging (S.S.V.), Agency for Science Technology and Research, 30 Medical Dr, Singapore 117609; Departments of Medicine (Y.M.K., J.G.E.), Obstetrics and Gynaecology (M.T.T., Y.S.C.), and Pediatrics (L.P.C.S., Y.S.L.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore (L.P.C.S., Y.S.L.); Departments of Pediatric Endocrinology (F.K.P.Y.), Obstetrics and Gynaecology (K.H.T.), and Diagnostic and Interventional Imaging (M.V.F.), KK Women's and Children's Hospital, Singapore; Pediatrics Academic Clinical Programme (F.K.P.Y.), Academic Medicine (K.H.T.), Duke-National University of Singapore Medical School, Singapore (F.K.P.Y., K.H.T.); Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (F.K.P.Y.); Medical Research Council Lifecourse Epidemiology Unit and National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, England (K.M.G.); School of Engineering, Pforzheim University, Pforzheim, Germany (U.C.M.); Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (J.G.E.); Folkhälsan Research Center, Helsinki, Finland (J.G.E.); and Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore (M.F.); Institute of Data Science, National University of Singapore, Singapore (M.F.)
| |
Collapse
|
38
|
Elkafrawy H, Mehanna R, Ali F, Barghash A, Dessouky I, Jernerén F, Turner C, Refsum H, Elshorbagy A. Extracellular cystine influences human preadipocyte differentiation and correlates with fat mass in healthy adults. Amino Acids 2021; 53:1623-1634. [PMID: 34519922 PMCID: PMC8521515 DOI: 10.1007/s00726-021-03071-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/19/2021] [Indexed: 02/08/2023]
Abstract
Plasma cysteine is associated with human obesity, but it is unknown whether this is mediated by reduced, disulfide (cystine and mixed-disulfides) or protein-bound (bCys) fractions. We investigated which cysteine fractions are associated with adiposity in vivo and if a relevant fraction influences human adipogenesis in vitro. In the current study, plasma cysteine fractions were correlated with body fat mass in 35 adults. Strong positive correlations with fat mass were observed for cystine and mixed disulfides (r ≥ 0.61, P < 0.001), but not the quantitatively major form, bCys. Primary human preadipocytes were differentiated in media containing cystine concentrations varying from 10-50 μM, a range similar to that in plasma. Increasing extracellular cystine (10-50 μM) enhanced mRNA expression of PPARG2 (to sixfold), PPARG1, PLIN1, SCD1 and CDO1 (P = 0.042- < 0.001). Adipocyte lipid accumulation and lipid-droplet size showed dose-dependent increases from lowest to highest cystine concentrations (P < 0.001), and the malonedialdehyde/total antioxidant capacity increased, suggesting increased oxidative stress. In conclusion, increased cystine concentrations, within the physiological range, are positively associated with both fat mass in healthy adults and human adipogenic differentiation in vitro. The potential role of cystine as a modifiable factor regulating human adipocyte turnover and metabolism deserves further study.
Collapse
Affiliation(s)
- Hagar Elkafrawy
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Radwa Mehanna
- Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Department of Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Fayrouz Ali
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ayman Barghash
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Iman Dessouky
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Fredrik Jernerén
- Department of Pharmacology, University of Oxford, Oxford, UK
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Cheryl Turner
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Helga Refsum
- Department of Pharmacology, University of Oxford, Oxford, UK
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Amany Elshorbagy
- Department of Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
- Department of Pharmacology, University of Oxford, Oxford, UK.
| |
Collapse
|
39
|
Brand T, van den Munckhof ICL, van der Graaf M, Schraa K, Dekker HM, Joosten LAB, Netea MG, Riksen NP, de Graaf J, Rutten JHW. Superficial vs Deep Subcutaneous Adipose Tissue: Sex-Specific Associations With Hepatic Steatosis and Metabolic Traits. J Clin Endocrinol Metab 2021; 106:e3881-e3889. [PMID: 34137897 PMCID: PMC8571813 DOI: 10.1210/clinem/dgab426] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Indexed: 01/21/2023]
Abstract
CONTEXT Subcutaneous adipose tissue (SAT) is not homogeneous, as the fascia scarpa separates the deep SAT (dSAT) from the superficial SAT (sSAT). OBJECTIVE The aim of this study is to evaluate the sex-specific associations of sSAT and dSAT with hepatic steatosis and metabolic syndrome in overweight individuals. METHODS We recruited 285 individuals with a body mass index (BMI) greater than or equal to 27 and aged 55 to 81 years. Abdominal magnetic resonance imaging was performed around level L4 to L5 to measure visceral adipose tissue (VAT), dSAT, and sSAT volumes. The amount of hepatic fat was quantified by MR spectroscopy. RESULTS Men had significantly higher volumes of VAT (122.6 cm3 vs 98.7 cm3, P < .001) and had only half the volume of sSAT compared to women adjusted for BMI (50.3 cm3 in men vs 97.0 cm3 in women, P < .001). dSAT correlated significantly with hepatic fat content in univariate analysis (standardized β = .190, P < .05), while VAT correlated significantly with hepatic steatosis in a multivariate model, adjusted for age, alcohol use, and other abdominal fat compartments (standardized β = .184, P = .037). Moreover, dSAT in men correlated negatively with HDL cholesterol (standardized β = -0.165, P = .038) in multivariate analyses. In women with a BMI between 30 and 40, in a multivariate model adjusted for age, alcohol use, and other abdominal fat compartments, VAT correlated positively (standardized β = -.404, P = .003), and sSAT negatively (standardized β = -.300, P = .04) with hepatic fat content. CONCLUSION In men, dSAT is associated with hepatic steatosis and adverse metabolic traits, such as lower HDL cholesterol levels, whereas in women with obesity sSAT shows a beneficial relation with respect to hepatic fat content.
Collapse
Affiliation(s)
- Tessa Brand
- Department of Internal Medicine, Division of Vascular Medicine 463, Radboud University Medical Center, HB Nijmegen, the Netherlands
| | | | - Marinette van der Graaf
- Department of Medical Imaging, Radboud University Medical Center, HB Nijmegen, the Netherlands
| | - Kiki Schraa
- Department of Internal Medicine, Division of Vascular Medicine 463, Radboud University Medical Center, HB Nijmegen, the Netherlands
| | - Helena Maria Dekker
- Department of Medical Imaging, Radboud University Medical Center, HB Nijmegen, the Netherlands
| | - Leonardus Antonius Bernardus Joosten
- Department of Internal Medicine, Division of Vascular Medicine 463, Radboud University Medical Center, HB Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai Gheorghe Netea
- Department of Internal Medicine, Division of Vascular Medicine 463, Radboud University Medical Center, HB Nijmegen, the Netherlands
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Niels Peter Riksen
- Department of Internal Medicine, Division of Vascular Medicine 463, Radboud University Medical Center, HB Nijmegen, the Netherlands
| | - Jacqueline de Graaf
- Department of Internal Medicine, Division of Vascular Medicine 463, Radboud University Medical Center, HB Nijmegen, the Netherlands
| | - Joseph Henricus Wilhelmus Rutten
- Department of Internal Medicine, Division of Vascular Medicine 463, Radboud University Medical Center, HB Nijmegen, the Netherlands
- Correspondence: J. H. W. Rutten, MD, PhD, Department of Internal Medicine (463), Radboudumc Nijmegen, P.O. Box 9101, 6500 HB Nijmegen, the Netherlands.
| |
Collapse
|
40
|
Teklu M, Zhou W, Kapoor P, Patel N, Playford MP, Sorokin AV, Dey AK, Teague HL, Manyak GA, Rodante JA, Keel A, Chen MY, Bluemke DA, Khera AV, Mehta NN. Abdominal subcutaneous adipose tissue negatively associates with subclinical coronary artery disease in men with psoriasis. Am J Prev Cardiol 2021; 8:100231. [PMID: 34553185 PMCID: PMC8441148 DOI: 10.1016/j.ajpc.2021.100231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/13/2021] [Accepted: 08/02/2021] [Indexed: 11/25/2022] Open
Abstract
Objective Understand the relationship between abdominal subcutaneous adipose tissue (ASAT) and coronary atherosclerosis defined as noncalcified and lipid-rich necrotic core burden in psoriasis. Methods We performed a cross-sectional study of 232 participants (92 women) with psoriasis and without known cardiovascular disease. Participants underwent coronary computed tomography angiography to characterize coronary atherosclerosis burden and low dose abdominal computed tomography to quantify subcutaneous and visceral adipose tissue. Fat depot volumes were first adjusted for each participant's BMI (ASATadjBMI). Results In women, there was a positive correlation between ASATadjBMI and systemic inflammation as assessed by hs-C-reactive protein (r=0.30; p=.004) and GlycA (r=0.29; p=.007) as well as total cholesterol (r=0.24; p=.02) and low-density lipoprotein cholesterol (r=0.22; p=.04). In men, ASATadjBMI correlated with hs-C-reactive protein (r=0.18; p=.04) and insulin resistance (r=0.17; p=.04). In models fully adjusted for traditional cardiovascular risk factors, ASATadjBMI negatively associated with noncalcified and lipid-rich necrotic core burden in men (β= -0.17; p=.03, β= -0.20; p=.03, respectively), but not women (β= -0.06; p=.57, β= 0.09; p=.49, respectively) with psoriasis. Conclusions For a given BMI, ASAT negatively associated with coronary atherosclerosis burden in male participants with psoriasis. The observed sex-specific effects warrant further study of ASAT in states of chronic inflammation.
Collapse
Affiliation(s)
- Meron Teklu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wunan Zhou
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Promita Kapoor
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nidhi Patel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Martin P Playford
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander V Sorokin
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amit K Dey
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heather L Teague
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Grigory A Manyak
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Justin A Rodante
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrew Keel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marcus Y Chen
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - David A Bluemke
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Amit V Khera
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
41
|
A novel conjunctive microenvironment derived from human subcutaneous adipose tissue contributes to physiology of its superficial layer. Stem Cell Res Ther 2021; 12:480. [PMID: 34454629 PMCID: PMC8399854 DOI: 10.1186/s13287-021-02554-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/22/2021] [Indexed: 11/23/2022] Open
Abstract
Background In human subcutaneous adipose tissue, the superficial fascia distinguishes superficial and deep microenvironments showing extensions called retinacula cutis. The superficial subcutaneous adipose tissue has been described as hyperplastic and the deep subcutaneous adipose tissue as inflammatory. However, few studies have described stromal-vascular fraction (SVF) content and adipose-derived stromal/stem cells (ASCs) behavior derived from superficial and deep subcutaneous adipose tissue. In this study, we analyzed a third conjunctive microenvironment: the retinacula cutis superficialis derived from superficial subcutaneous adipose tissue. Methods The samples of abdominal human subcutaneous adipose tissue were obtained during plastic aesthetic surgery in France (Declaration DC-2008-162) and Brazil (Protocol 145/09). Results The SVF content was characterized in situ by immunofluorescence and ex vivo by flow cytometry revealing a high content of pre-adipocytes rather in superficial subcutaneous adipose tissue microenvironment. Adipogenic assays revealed higher percentage of lipid accumulation area in ASCs from superficial subcutaneous adipose tissue compared with retinacula cutis superficialis (p < 0.0001) and deep subcutaneous adipose tissue (p < 0.0001). The high adipogenic potential of superficial subcutaneous adipose tissue was corroborated by an up-regulation of adipocyte fatty acid-binding protein (FABP4) compared with retinacula cutis superficialis (p < 0.0001) and deep subcutaneous adipose tissue (p < 0.0001) and of C/EBPα (CCAAT/enhancer-binding protein alpha) compared with retinacula cutis superficialis (p < 0.0001) and deep subcutaneous adipose tissue (p < 0.0001) microenvironments. Curiously, ASCs from retinacula cutis superficialis showed a higher level of adiponectin receptor gene compared with superficial subcutaneous adipose tissue (p = 0.0409), widely known as an anti-inflammatory hormone. Non-induced ASCs from retinacula cutis superficialis showed higher secretion of human vascular endothelial growth factor (VEGF), compared with superficial (p = 0.0485) and deep (p = 0.0112) subcutaneous adipose tissue and with adipogenic-induced ASCs from superficial (p = 0.0175) and deep (p = 0.0328) subcutaneous adipose tissue. Furthermore, ASCs from retinacula cutis superficialis showed higher secretion of Chemokine (C–C motif) ligand 5 (CCL5) compared with non-induced (p = 0.0029) and induced (p = 0.0089) superficial subcutaneous adipose tissue. Conclusions This study highlights the contribution to ASCs from retinacula cutis superficialis in their angiogenic property previously described for the whole superficial subcutaneous adipose tissue besides supporting its adipogenic potential for superficial subcutaneous adipose tissue.
Collapse
|
42
|
Kałużna M, Czlapka-Matyasik M, Bykowska-Derda A, Moczko J, Ruchala M, Ziemnicka K. Indirect Predictors of Visceral Adipose Tissue in Women with Polycystic Ovary Syndrome: A Comparison of Methods. Nutrients 2021; 13:2494. [PMID: 34444654 PMCID: PMC8401513 DOI: 10.3390/nu13082494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/05/2021] [Accepted: 07/20/2021] [Indexed: 12/31/2022] Open
Abstract
Visceral adipose tissue (VAT) accumulation, is a part of a polycystic ovary syndrome (PCOS) phenotype. Dual-energy x-ray absorptiometry (DXA) provides a gold standard measurement of VAT. This study aimed to compare ten different indirect methods of VAT estimation in PCOS women. The study included 154 PCOS and 68 age- and BMI-matched control women. Subjects were divided into age groups: 18-30 y.o. and 30-40 y.o. Analysis included: body mass index (BMI), waist circumference (WC), waist-to-hip ratio (WHR), waist-to-height ratio (WHtR), waist/height 0.5 (WHT.5R), visceral adipose index (VAI), lipid accumulation product (LAP), and fat mass index (FMI). VAT accumulation, android-to-gynoid ratio (A/G), and total body fat (TBF) was measured by DXA. ROC analysis revealed that WHtR, WHT.5R, WC, BMI, and LAP demonstrated the highest predictive value in identifying VAT in the PCOS group. Lower cut-off values of BMI (23.43 kg/m2) and WHtR (0.45) were determined in the younger PCOS group and higher thresholds of WHtR (0.52) in the older PCOS group than commonly used. Measuring either: WHtR, WHT.5R, WC, BMI, or LAP, could help identify a subgroup of PCOS patients at high cardiometabolic risk. The current observations reinforce the importance of using special cut-offs to identify VAT, dependent on age and PCOS presence.
Collapse
Affiliation(s)
- Małgorzata Kałużna
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (M.R.); (K.Z.)
| | - Magdalena Czlapka-Matyasik
- Department of Human Nutrition and Dietetics, Poznan University of Life Sciences, 60-624 Poznan, Poland; (M.C.-M.); (A.B.-D.)
| | - Aleksandra Bykowska-Derda
- Department of Human Nutrition and Dietetics, Poznan University of Life Sciences, 60-624 Poznan, Poland; (M.C.-M.); (A.B.-D.)
| | - Jerzy Moczko
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
| | - Marek Ruchala
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (M.R.); (K.Z.)
| | - Katarzyna Ziemnicka
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (M.R.); (K.Z.)
| |
Collapse
|
43
|
Stein E, Barbiero S, Lucia Portal V, Luz VD, Marcadenti A. Association between Deep Subcutaneous Adipose Tissue Estimated by DAAT Index and Dietary Intake in Patients with Acute Coronary Syndrome. INTERNATIONAL JOURNAL OF CARDIOVASCULAR SCIENCES 2021. [DOI: 10.36660/ijcs.20200334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
44
|
Li M, Qian M, Kyler K, Xu J. Adipose Tissue-Endothelial Cell Interactions in Obesity-Induced Endothelial Dysfunction. Front Cardiovasc Med 2021; 8:681581. [PMID: 34277732 PMCID: PMC8282205 DOI: 10.3389/fcvm.2021.681581] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022] Open
Abstract
Obesity has a strong impact on the pathogenesis of cardiovascular disease, which raises enthusiasm to understand how excess adiposity causes vascular injury. Adipose tissue is an essential regulator of cardiovascular system through its endocrine and paracrine bioactive products. Obesity induces endothelial dysfunction, which often precedes and leads to the development of cardiovascular diseases. Connecting adipose tissue-endothelial cell interplay to endothelial dysfunction may help us to better understand obesity-induced cardiovascular disease. This Mini Review discussed (1) the general interactions and obesity-induced endothelial dysfunction, (2) potential targets, and (3) the outstanding questions for future research.
Collapse
Affiliation(s)
- Manna Li
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Ming Qian
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Kathy Kyler
- Office of Research Administration, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Jian Xu
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| |
Collapse
|
45
|
Abstract
Obese heart failure with preserved ejection fraction (HFpEF) is a distinct HFpEF phenotype. Sodium retention, high circulating neurohormone levels, alterations in energy substrate metabolism, group 3 pulmonary hypertension, pericardial restraint, and systemic inflammation are central pathophysiologic mechanisms. Confirming the diagnosis may be challenging and high suspicion is required. Reduction of visceral adipose tissue, via caloric restriction and/or bariatric surgery, may improve outcomes in obese HFpEF patients. Furthermore, mineralocorticoid receptor inhibition, neprilysin inhibition, and sodium-glucose cotransporter 2 inhibition can ameliorate the effects of adiposity on the cardiovascular system, allowing for promising new treatment targets for the obese HFpEF phenotype.
Collapse
Affiliation(s)
- Efstratios Koutroumpakis
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center, 6431 Fannin Street, MSB 1.220, Houston, TX 77030, USA
| | - Ramanjit Kaur
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center, 6431 Fannin Street, MSB 1.220, Houston, TX 77030, USA
| | - Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center, 6431 Fannin Street, MSB 1.220, Houston, TX 77030, USA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
46
|
Bae J, Ju JW, Lee S, Nam K, Kim TK, Jeon Y, Cho YJ. Association between abdominal fat and mortality in patients undergoing cardiovascular surgery. Ann Thorac Surg 2021; 113:1506-1513. [PMID: 34116000 DOI: 10.1016/j.athoracsur.2021.05.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 05/09/2021] [Accepted: 05/14/2021] [Indexed: 11/01/2022]
Abstract
BACKGROUND Obesity is associated with reduced postoperative mortality in patients undergoing cardiovascular surgery. However, body mass index cannot differentiate abdominal fat composition. This study evaluated the relationships between total abdominal, subcutaneous, and visceral fat composition and postoperative mortality in East Asian patients undergoing cardiovascular surgery. METHODS Adult patients who underwent cardiovascular surgery between October 2004 and December 2016 were retrospectively included. Total, subcutaneous and visceral fat areas were measured from cross-sectional computed tomography images. The relationships between each fat composition and mortality were evaluated. RESULTS A total of 3,661 patients were analyzed, and overall mortality was 19.9% (729 died) during the 4.6 year median follow-up period. The risks of all-cause and cardiac-cause mortality decreased as subcutaneous fat composition increased (adjusted hazard ratio [aHR], 0.997; 95% confidence interval [CI], 0.994-1.000 and aHR, 0.994; 95% CI, 0.989-0.999; P = .02 and = .01, respectively). No association was detected between the total and visceral fat area and mortality. CONCLUSIONS Reduced abdominal subcutaneous fat, but not the total or visceral fat composition was associated with higher all-cause and cardiac-cause mortality after cardiovascular surgery in East Asian patients, consisting mainly of normal or overweight patients.
Collapse
Affiliation(s)
- Jinyoung Bae
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Jae-Woo Ju
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Seohee Lee
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Karam Nam
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Tae Kyong Kim
- Department of Anesthesiology and Pain Medicine, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Yunseok Jeon
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Youn Joung Cho
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
47
|
Jang HY, Han Y, Yoo HJ, Lee JH, Kim M. Effects of short-term dietary restriction on plasma metabolites and the subcutaneous fat area according to metabolic status in obese individuals: a case-control study. Diabetol Metab Syndr 2021; 13:62. [PMID: 34099056 PMCID: PMC8186103 DOI: 10.1186/s13098-021-00679-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/26/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Research elucidating the metabolic mechanisms that differentiate subtypes of obesity has been increasing. We aimed to investigate the effects of a 12-week dietary intervention on the metabolomic profiles of obese subjects. METHODS Subjects followed a 12-week dietary restriction protocol consisting of a 300 kcal/day reduction in their usual caloric intake. Twenty-nine obese subjects were included and divided into two groups: the metabolic status maintenance group (n = 17, controls) and the metabolic status improvement group (n = 12, tests). We analyzed the somatometric and biochemical parameters and performed ultra-performance liquid chromatography-mass spectrometry analysis of the plasma metabolites. RESULTS At 12 weeks, the fat percentage, whole fat area (WFA), subcutaneous fat area (SFA) at the L1 vertebra, and the levels of triglycerides, gamma-glutamyltransferase (gamma-GT), and leptin were markedly decreased in the metabolic status improvement group, while the level of high-density lipoprotein cholesterol increased compared with that in the metabolic status maintenance group. Metabolomic profiling at 12 weeks showed substantial differences in 4-aminobutyraldehyde (p = 0.005) and 4'-apo-β-carotenal (p = 0.024) between the two groups. Furthermore, an AUC value of 0.89 was obtained for the following seven featured biomarkers: triglycerides, gamma-GT, leptin, fat percentage, WFA, and SFA at the L1 vertebra, and 4-aminobutyraldehyde. CONCLUSIONS We demonstrated that 4-aminobutyraldehyde and related regional fat distribution parameters were strongly associated with obesity according to metabolic status. Thus, these biomarkers are potentially valuable in confirming the efficacy of short-term interventions and predicting metabolic status in obese individuals. TRIALS REGISTRATION This study was registered at ClinicalTrials.gov under NCT03135132 (registered 1 May 2017-retrospectively registered).
Collapse
Affiliation(s)
- Hye Yoon Jang
- Department of Science for Aging, Graduate School of Yonsei University, Seoul, 03722, Korea
| | - Youngmin Han
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
| | - Hye Jin Yoo
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, 03722, Korea
| | - Jong Ho Lee
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, 03722, Korea
| | - Minjoo Kim
- Department of Food and Nutrition, College of Life Science and Nano Technology, Hannam University, Daejeon, 34054, Korea.
| |
Collapse
|
48
|
Ding L, Fan Y, He J, Wang R, He Q, Cui J, Ma Z, Zheng F, Gao H, Dai C, Wei H, Li J, Cao Y, Hu G, Liu M. Different Indicators of Adiposity and Fat Distribution and Cardiometabolic Risk Factors in Patients with Type 2 Diabetes. Obesity (Silver Spring) 2021; 29:837-845. [PMID: 33899339 PMCID: PMC9115840 DOI: 10.1002/oby.23151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/11/2021] [Accepted: 02/05/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE This study aimed to evaluate the effect of adiposity and fat distribution on the odds of elevated cardiovascular risk factors among adults with type 2 diabetes mellitus. METHODS The present cross-sectional study included 2,427 adults with type 2 diabetes mellitus. Body fat was assessed by dual-energy x-ray absorptiometry. Multivariate-adjusted logistic regression was used to estimate effects of adiposity parameters on elevated hemoglobin A1c (HbA1c , ≥7.0%), hypertension (blood pressure ≥140/90 mmHg), and elevated low-density lipoprotein (LDL) cholesterol (≥2.6 mmol/L). RESULTS The multivariable-adjusted odds ratio (OR) for elevated HbA1c was 0.82 (95% CI: 0.70-0.96) for each SD increase in leg fat mass. The multivariable-adjusted OR for hypertension was 1.15 (95% CI: 1.00-1.32) for each SD increase in android fat mass. Multivariable-adjusted ORs for elevated LDL cholesterol ranged from 1.16 (95% CI: 1.00-1.35) to 1.27 (95% CI: 1.06-1.51) for each SD increase in arm and android fat mass and percentage of total, truncal, arm, and android fat. Each SD increase in BMI, truncal-to-leg fat ratio, and android-to-gynoid fat ratio was significantly associated with increased risks of elevated HbA1c , hypertension, and elevated LDL cholesterol. CONCLUSIONS Subcutaneous fat in the lower body was associated with a more favorable glycemic profile, but not blood pressure or lipid profile, whereas central adiposity was associated with poor control of cardiovascular risk factors among patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Li Ding
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuxin Fan
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Jing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Ruodan Wang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingqiu Cui
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhongshu Ma
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Fangqiu Zheng
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Hua Gao
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Chenlin Dai
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyan Wei
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuming Cao
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
49
|
Porro S, Genchi VA, Cignarelli A, Natalicchio A, Laviola L, Giorgino F, Perrini S. Dysmetabolic adipose tissue in obesity: morphological and functional characteristics of adipose stem cells and mature adipocytes in healthy and unhealthy obese subjects. J Endocrinol Invest 2021; 44:921-941. [PMID: 33145726 DOI: 10.1007/s40618-020-01446-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
The way by which subcutaneous adipose tissue (SAT) expands and undergoes remodeling by storing excess lipids through expansion of adipocytes (hypertrophy) or recruitment of new precursor cells (hyperplasia) impacts the risk of developing cardiometabolic and respiratory diseases. In unhealthy obese subjects, insulin resistance, type 2 diabetes, hypertension, and obstructive sleep apnoea are typically associated with pathologic SAT remodeling characterized by adipocyte hypertrophy, as well as chronic inflammation, hypoxia, increased visceral adipose tissue (VAT), and fatty liver. In contrast, metabolically healthy obese individuals are generally associated with SAT development characterized by the presence of smaller and numerous mature adipocytes, and a lower degree of VAT inflammation and ectopic fat accumulation. The remodeling of SAT and VAT is under genetic regulation and influenced by inherent depot-specific differences of adipose tissue-derived stem cells (ASCs). ASCs have multiple functions such as cell renewal, adipogenic capacity, and angiogenic properties, and secrete a variety of bioactive molecules involved in vascular and extracellular matrix remodeling. Understanding the mechanisms regulating the proliferative and adipogenic capacity of ASCs from SAT and VAT in response to excess calorie intake has become a focus of interest over recent decades. Here, we summarize current knowledge about the biological mechanisms able to foster or impair the recruitment and adipogenic differentiation of ASCs during SAT and VAT development, which regulate body fat distribution and favorable or unfavorable metabolic responses.
Collapse
Affiliation(s)
- S Porro
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - V A Genchi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Cignarelli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Natalicchio
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - L Laviola
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - F Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy.
| | - S Perrini
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
50
|
Association of Body Composition with Type 2 Diabetes: A Retrospective Chart Review Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18094421. [PMID: 33919339 PMCID: PMC8122668 DOI: 10.3390/ijerph18094421] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/25/2022]
Abstract
This study analyzed the body composition of individuals with type 2 diabetes (T2DM). In this retrospective chart review study, body composition was measured through multifrequency bioelectrical impedance analysis (InBody 770). Body composition assessments were conducted in individuals with T2DM, who were aged ≥18 years. The parameters included body mass index (BMI), body fat mass (BFM), fat-free mass (FFM), visceral fat area, percent body fat (PBF), appendicular skeletal muscle mass (ASM), and skeletal muscle index (SMI). One-way ANOVA and independent t-tests were used to calculate differences in body composition distribution by age and sex. A total of 2404 participants were recruited. The prevalence of overall low muscle mass and sarcopenic obesity was 28.0% and 18.7%, respectively, which increased with age. The overall prevalence of obesity when PBF was used was 71.5%, which was higher than that when BMI was applied (32.4%). The normal BMI group exhibited a prevalence of low muscle mass of 55.6% and sarcopenic obesity of 34.8%. For both men and women, bodyweight, BFM, FFM, ASM, and SMI all decreased with age. The prevalence of low muscle mass and sarcopenic obesity was high in older adults and people with normal BMI. Using BMI to assess obesity and determine insufficient muscle mass underestimates the prevalence of obesity and neglects the problems of sarcopenia and high body fat in people with normal BMI.
Collapse
|