1
|
Fortes YM, Souza-Gomes AF, Moreira ARS, Campos LN, de Moura SS, Barroso LSS, de Faria MHS, de Barros Fernandes H, de Miranda AS, Martins-Costa HC, Simões e Silva AC, Moreira JM, Nunes-Silva A. A single session of strength training changed plasma levels of resistin, but not leptin in overweight and obese men. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:324-330. [PMID: 39309458 PMCID: PMC11411330 DOI: 10.1016/j.smhs.2023.12.001] [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: 06/08/2023] [Revised: 11/20/2023] [Accepted: 12/04/2023] [Indexed: 09/25/2024] Open
Abstract
Obesity has a complex multifactorial etiology and is characterized by excessive accumulation of adipose tissue. Visceral adipose tissue has deleterious effects on health because it secretes large amounts of inflammatory cytokines. Nutritional calorie restriction associated with strength training may be useful in managing chronic systemic inflammation. This study aimed to evaluate the acute effect of a single strength-training session on plasma adipokine levels in sedentary, overweight, and obese young men. This study included twelve men (Age: [34.95 ± 9.77] years; Height: [174.16 ± 3.66] centimeter [cm]; Weight: [97.83 ± 12.87] kilogram (kg); body mass index [BMI]: [32.30 ± 4.51] kg/m2), who performed a single strength training session. The strength training protocol consisted of 4 sets of 12 repetitions in the following six exercises, 45° leg press, bench press, leg extension, machine row, leg curl, and shoulder press. Blood samples were collected before, immediately after, and 1-h subsequent after strength training. The plasma levels of resistin and leptin were measured. A significant decrease in resistin levels were found 1 h after the strength training session if compared to levels before the training session (pre-[before] [2 390 ± 1 199] picograms per milliliter [pg/mL] vs post-1 h [1-h subsequent] [1 523 ± 798],6 pg/mL, p = 0.002 8). The plasma leptin levels did not differ at any time point. In conclusion, a very well controlled single session of strength training significantly decreased the plasma levels of resistin without altering the concentration of leptin in overweight and obese individuals. This effect, at least in part, supports the benefits of exercise by reducing the low grade inflammation and insulin resistance in obesity.
Collapse
Affiliation(s)
- Yago Martins Fortes
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Postgraduate Program in Health and Nutrition, Nutrition School, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Antonio Felipe Souza-Gomes
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Postgraduate Program in Health and Nutrition, Nutrition School, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Alessandro Roberto Silveira Moreira
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Postgraduate Program in Health and Nutrition, Nutrition School, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Leo Nogueira Campos
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Samara Silva de Moura
- Postgraduate Program in Health and Nutrition, Nutrition School, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Lucélia Scarabeli Silva Barroso
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Interdisciplinary Medical Research Laboratory, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcelo Henrique Salviano de Faria
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Interdisciplinary Medical Research Laboratory, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Heliana de Barros Fernandes
- Neurobiology Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aline Silva de Miranda
- Neurobiology Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Ana Cristina Simões e Silva
- Interdisciplinary Medical Research Laboratory, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Janaina Matos Moreira
- Department of Pediatrics, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Albená Nunes-Silva
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Postgraduate Program in Health and Nutrition, Nutrition School, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| |
Collapse
|
2
|
Wang S, Yin J, Liu Z, Liu X, Tian G, Xin X, Qin Y, Feng X. Metabolic disorders, inter-organ crosstalk, and inflammation in the progression of metabolic dysfunction-associated steatotic liver disease. Life Sci 2024; 359:123211. [PMID: 39491769 DOI: 10.1016/j.lfs.2024.123211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/20/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a global health concern, affecting over 30 % of adults. It is a principal driver in the development of cirrhosis and hepatocellular carcinoma. The complex pathogenesis of MASLD involves an excessive accumulation of lipids, subsequently disrupting lipid metabolism and prompting inflammation within the liver. This review synthesizes the recent research progress in understanding the mechanisms contributing to MASLD progression, with particular emphasis on metabolic disorders and interorgan crosstalk. We highlight the molecular mechanisms linked to these factors and explore their potential as novel targets for pharmacological intervention. The insights gleaned from this article have important implications for both the prevention and therapeutic management of MASLD.
Collapse
Affiliation(s)
- Shendong Wang
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Zhaojun Liu
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Xin Liu
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Ge Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
| | - Xijian Xin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Yiming Qin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China.
| |
Collapse
|
3
|
Contessi Negrini N, Pellegrinelli V, Salem V, Celiz A, Vidal-Puig A. Breaking barriers in obesity research: 3D models of dysfunctional adipose tissue. Trends Biotechnol 2024:S0167-7799(24)00278-6. [PMID: 39443224 DOI: 10.1016/j.tibtech.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Obesity is a global health crisis characterised by excessive accumulation of adipose tissue (AT). Under obesogenic conditions, this metabolically active tissue undergoes fibrosis and inflammation, leading to obesity-linked comorbidities. Modelling AT is essential for understanding its pathophysiology and developing treatments to protect against metabolic complications. 3D in vitro AT models are promising tools that address the limitations of traditional 2D in vitro models and in vivo animal models, providing enhanced biomimetic and human-relevant platforms. 3D models facilitate the study of AT pathophysiology and therapeutic screening. This review discusses the crucial role of AT in obesity-linked comorbidities, its dynamicity and complexity, and recent advances in engineering 3D scaffold-based in vitro dysfunctional AT models, highlighting potential breakthroughs in metabolic research and beyond.
Collapse
Affiliation(s)
- Nicola Contessi Negrini
- Department of Bioengineering, Imperial College London, London, UK; The Francis Crick Institute, London, UK.
| | | | - Victoria Salem
- Department of Bioengineering, Imperial College London, London, UK
| | - Adam Celiz
- Department of Bioengineering, Imperial College London, London, UK; The Francis Crick Institute, London, UK
| | - Antonio Vidal-Puig
- MRC Institute of Metabolic Science and Medical Research Council, Cambridge, UK; Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, PR China; Centro de Investigacion Principe Felipe (CIPF), Valencia, Spain; Cambridge Heart and Lung Research Institute, Cambridge, UK
| |
Collapse
|
4
|
Dronkers J, van Veldhuisen DJ, van der Meer P, Meems LMG. Heart Failure and Obesity: Unraveling Molecular Mechanisms of Excess Adipose Tissue. J Am Coll Cardiol 2024; 84:1666-1677. [PMID: 39415402 DOI: 10.1016/j.jacc.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/01/2024] [Accepted: 07/11/2024] [Indexed: 10/18/2024]
Abstract
Obesity is an ongoing pandemic and is associated with the development of heart failure (HF), and especially HF with preserved ejection fraction. The definition of obesity is currently based on anthropometric measurements but neglects the location and molecular properties of excess fat. Important depots associated with HF development are subcutaneous adipose tissue and visceral adipose tissue, both located in the abdominal region, and epicardial adipose tissue (EAT) surrounding the myocardium. However, mechanisms linking these different adipose tissue depots to HF development are incompletely understood. EAT in particular is of great interest because of its close proximity to the heart. In this review, we therefore focus on the characteristics of different adipose tissue depots and their response to obesity. In addition, we evaluate how different mechanisms associated with EAT expansion potentially contribute to HF and in particular HF with preserved ejection fraction development.
Collapse
Affiliation(s)
- Just Dronkers
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Dirk J van Veldhuisen
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Peter van der Meer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Laura M G Meems
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands.
| |
Collapse
|
5
|
Jornayvaz FR, Gariani K, Somm E, Jaquet V, Bouzakri K, Szanto I. NADPH oxidases in healthy white adipose tissue and in obesity: function, regulation, and clinical implications. Obesity (Silver Spring) 2024; 32:1799-1811. [PMID: 39315402 DOI: 10.1002/oby.24113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/19/2024] [Accepted: 06/11/2024] [Indexed: 09/25/2024]
Abstract
Reactive oxygen species, when produced in a controlled manner, are physiological modulators of healthy white adipose tissue (WAT) expansion and metabolic function. By contrast, unbridled production of oxidants is associated with pathological WAT expansion and the establishment of metabolic dysfunctions, most notably insulin resistance and type 2 diabetes mellitus. NADPH oxidases (NOXs) produce oxidants in an orderly fashion and are present in adipocytes and in other diverse WAT-constituent cell types. Recent studies have established several links between aberrant NOX-derived oxidant production, adiposity, and metabolic homeostasis. The objective of this review is to highlight the physiological roles attributed to diverse NOX isoforms in healthy WAT and summarize current knowledge of the metabolic consequences related to perturbations in their adequate oxidant production. We detail WAT-related alterations in preclinical investigations conducted in NOX-deficient murine models. In addition, we review clinical studies that have employed NOX inhibitors and currently available data related to human NOX mutations in metabolic disturbances. Future investigations aimed at understanding the integration of NOX-derived oxidants in the regulation of the WAT cellular redox network are essential for designing successful redox-related precision therapies to curb obesity and attenuate obesity-associated metabolic pathologies.
Collapse
Affiliation(s)
- François R Jornayvaz
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Karim Gariani
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Emmanuel Somm
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Vincent Jaquet
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
- RE.A.D.S. Unit (Readers, Assay Development and Screening Unit), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Karim Bouzakri
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Ildiko Szanto
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
6
|
Di Rocco G, Trivisonno A, Trivisonno G, Toietta G. Dissecting human adipose tissue heterogeneity using single-cell omics technologies. Stem Cell Res Ther 2024; 15:322. [PMID: 39334440 PMCID: PMC11437900 DOI: 10.1186/s13287-024-03931-w] [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: 07/04/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Single-cell omics technologies that profile genes (genomic and epigenomic) and determine the abundance of mRNA (transcriptomic), protein (proteomic and secretomic), lipids (lipidomic), and extracellular matrix (matrisomic) support the dissection of adipose tissue heterogeneity at unprecedented resolution in a temporally and spatially defined manner. In particular, cell omics technologies may provide innovative biomarkers for the identification of rare specific progenitor cell subpopulations, assess transcriptional and proteomic changes affecting cell proliferation and immunomodulatory potential, and accurately define the lineage hierarchy and differentiation status of progenitor cells. Unraveling adipose tissue complexity may also provide for the precise assessment of a dysfunctional state, which has been associated with cancer, as cancer-associated adipocytes play an important role in shaping the tumor microenvironment supporting tumor progression and metastasis, obesity, metabolic syndrome, and type 2 diabetes mellitus. The information collected by single-cell omics has relevant implications for regenerative medicine because adipose tissue is an accessible source of multipotent cells; alternative cell-free approaches, including the use of adipose tissue stromal cell-conditioned medium, extracellular vesicles, or decellularized extracellular matrix, are clinically valid options. Subcutaneous white adipose tissue, which is generally harvested via liposuction, is highly heterogeneous because of intrinsic biological variability and extrinsic inconsistencies in the harvesting and processing procedures. The current limited understanding of adipose tissue heterogeneity impinges on the definition of quality standards appropriate for clinical translation, which requires consistency and uniformity of the administered product. We review the methods used for dissecting adipose tissue heterogeneity and provide an overview of advances in omics technology that may contribute to the exploration of heterogeneity and dynamics of adipose tissue at the single-cell level.
Collapse
Affiliation(s)
- Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Angelo Trivisonno
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | | | - Gabriele Toietta
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi, 53, 00144, Rome, Italy.
| |
Collapse
|
7
|
Vyas V, Sandhar B, Keane JM, Wood EG, Blythe H, Jones A, Shahaj E, Fanti S, Williams J, Metic N, Efremova M, Ng HL, Nageswaran G, Byrne S, Feldhahn N, Marelli-Berg F, Chain B, Tinker A, Finlay MC, Longhi MP. Tissue-resident memory T cells in epicardial adipose tissue comprise transcriptionally distinct subsets that are modulated in atrial fibrillation. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1067-1082. [PMID: 39271815 PMCID: PMC11399095 DOI: 10.1038/s44161-024-00532-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 07/29/2024] [Indexed: 09/15/2024]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia and carries an increased risk of stroke and heart failure. Here we investigated how the immune infiltrate of human epicardial adipose tissue (EAT), which directly overlies the myocardium, contributes to AF. Flow cytometry analysis revealed an enrichment of tissue-resident memory T (TRM) cells in patients with AF. Cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) and single-cell T cell receptor (TCR) sequencing identified two transcriptionally distinct CD8+ TRM cells that are modulated in AF. Spatial transcriptomic analysis of EAT and atrial tissue identified the border region between the tissues to be a region of intense inflammatory and fibrotic activity, and the addition of TRM populations to atrial cardiomyocytes demonstrated their ability to differentially alter calcium flux as well as activate inflammatory and apoptotic signaling pathways. This study identified EAT as a reservoir of TRM cells that can directly modulate vulnerability to cardiac arrhythmia.
Collapse
Affiliation(s)
- Vishal Vyas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, London, UK
| | - Balraj Sandhar
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jack M Keane
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Elizabeth G Wood
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Hazel Blythe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Aled Jones
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Eriomina Shahaj
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Silvia Fanti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jack Williams
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nasrine Metic
- Cancer Research UK, Barts Centre, Queen Mary University of London, London, UK
| | - Mirjana Efremova
- Cancer Research UK, Barts Centre, Queen Mary University of London, London, UK
| | - Han Leng Ng
- Department of Immunology and Inflammation, Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| | - Gayathri Nageswaran
- UCL Division of Infection and Immunity, University College London, London, UK
| | - Suzanne Byrne
- UCL Division of Infection and Immunity, University College London, London, UK
| | - Niklas Feldhahn
- Department of Immunology and Inflammation, Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Benny Chain
- UCL Division of Infection and Immunity, University College London, London, UK
| | - Andrew Tinker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Malcolm C Finlay
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, London, UK
| | - M Paula Longhi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
8
|
Kostecka A, Kalamon N, Skoniecka A, Koczkowska M, Skowron PM, Piotrowski A, Pikuła M. Adipose-derived mesenchymal stromal cells in clinical trials: Insights from single-cell studies. Life Sci 2024; 351:122761. [PMID: 38866216 DOI: 10.1016/j.lfs.2024.122761] [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: 04/02/2024] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Mesenchymal Stromal Cells (MSCs) offer tremendous potential for the treatment of various diseases and their healing properties have been explored in hundreds of clinical trials. These trails primarily focus on immunological and neurological disorders, as well as regenerative medicine. Adipose tissue is a rich source of mesenchymal stromal cells and methods to obtain and culture adipose-derived MSCs (AD-MSCs) have been well established. Promising results from pre-clinical testing of AD-MSCs activity prompted clinical trials that further led to the approval of AD-MSCs for the treatment of complex perianal fistulas in Crohn's disease and subcutaneous tissue defects. However, AD-MSC heterogeneity along with various manufacturing protocols or different strategies to boost their activity create the need for standardized quality control procedures and safety assessment of the intended cell product. High-resolution transcriptomic methods have been recently gaining attention, as they deliver insight into gene expression profiles of individual cells, helping to deconstruct cellular hierarchy and differentiation trajectories, and to understand cell-cell interactions within tissues. This article presents a comprehensive overview of completed clinical trials evaluating the safety and efficacy of AD-MSC treatment, together with current single-cell studies of human AD-MSC. Furthermore, our work emphasizes the increasing significance of single-cell research in elucidating the mechanisms of cellular action and predicting their therapeutic effects.
Collapse
Affiliation(s)
- Anna Kostecka
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Natalia Kalamon
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland.
| | - Aneta Skoniecka
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, 80-211 Gdańsk, Poland.
| | - Magdalena Koczkowska
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland.
| | - Arkadiusz Piotrowski
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, 80-211 Gdańsk, Poland.
| |
Collapse
|
9
|
Jang S, Hong W, Moon Y. Obesity-compromised immunity in post-COVID-19 condition: a critical control point of chronicity. Front Immunol 2024; 15:1433531. [PMID: 39188722 PMCID: PMC11345197 DOI: 10.3389/fimmu.2024.1433531] [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: 05/16/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Post-COVID-19 condition is recognized as a multifactorial disorder, with persistent presence of viral antigens, discordant immunity, delayed viral clearance, and chronic inflammation. Obesity has emerged as an independent risk factor for both SARS-CoV-2 infection and its subsequent sequelae. In this study, we aimed to predict the molecular mechanisms linking obesity and post-COVID-19 distress. Viral antigen-exposed adipose tissues display remarkable levels of viral receptors, facilitating viral entry, deposition, and chronic release of inflammatory mediators and cells in patients. Subsequently, obesity-associated inflammatory insults are predicted to disturb cellular and humoral immunity by triggering abnormal cell differentiation and lymphocyte exhaustion. In particular, the decline in SARS-CoV-2 antibody titers and T-cell exhaustion due to chronic inflammation may account for delayed virus clearance and persistent activation of inflammatory responses. Taken together, obesity-associated defective immunity is a critical control point of intervention against post-COVID-19 progression, particularly in subjects with chronic metabolic distress.
Collapse
Affiliation(s)
- Soonwoo Jang
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
| | - Wooyoung Hong
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
10
|
Yang S, Sun Y, Yan C. Recent advances in the use of extracellular vesicles from adipose-derived stem cells for regenerative medical therapeutics. J Nanobiotechnology 2024; 22:316. [PMID: 38844939 PMCID: PMC11157933 DOI: 10.1186/s12951-024-02603-4] [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: 03/21/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) are a subset of mesenchymal stem cells (MSCs) isolated from adipose tissue. They possess remarkable properties, including multipotency, self-renewal, and easy clinical availability. ADSCs are also capable of promoting tissue regeneration through the secretion of various cytokines, factors, and extracellular vesicles (EVs). ADSC-derived EVs (ADSC-EVs) act as intercellular signaling mediators that encapsulate a range of biomolecules. These EVs have been found to mediate the therapeutic activities of donor cells by promoting the proliferation and migration of effector cells, facilitating angiogenesis, modulating immunity, and performing other specific functions in different tissues. Compared to the donor cells themselves, ADSC-EVs offer advantages such as fewer safety concerns and more convenient transportation and storage for clinical application. As a result, these EVs have received significant attention as cell-free therapeutic agents with potential future application in regenerative medicine. In this review, we focus on recent research progress regarding regenerative medical use of ADSC-EVs across various medical conditions, including wound healing, chronic limb ischemia, angiogenesis, myocardial infarction, diabetic nephropathy, fat graft survival, bone regeneration, cartilage regeneration, tendinopathy and tendon healing, peripheral nerve regeneration, and acute lung injury, among others. We also discuss the underlying mechanisms responsible for inducing these therapeutic effects. We believe that deciphering the biological properties, therapeutic effects, and underlying mechanisms associated with ADSC-EVs will provide a foundation for developing a novel therapeutic approach in regenerative medicine.
Collapse
Affiliation(s)
- Song Yang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Yiran Sun
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China.
| | - Chenchen Yan
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| |
Collapse
|
11
|
Grodsky L, Wilson M, Rathinasabapathy T, Komarnytsky S. Triptolide Administration Alters Immune Responses to Mitigate Insulin Resistance in Obese States. Biomolecules 2024; 14:395. [PMID: 38672413 PMCID: PMC11048574 DOI: 10.3390/biom14040395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Individuals who are overweight or obese are at increased risk of developing prediabetes and type 2 diabetes, yet the direct molecular mechanisms that connect diabetes to obesity are not clear. Chronic, sustained inflammation is considered a strong risk factor in these interactions, directed in part by the short-lived gene expression programs encoding for cytokines and pro-inflammatory mediators. In this study, we show that triptolide administration in the C57BL/6 diet-induced obese mice at up to 10 μg/kg/day for 10 weeks attenuated the development of insulin resistance and diabetes, but not obesity, in these animals. Significant reductions in adipose tissue inflammation and improved insulin sensitivity were observed in the absence of changes in food intake, body weight, body composition, or energy expenditure. Analysis of the core cluster of biomarkers that drives pro-inflammatory responses in the metabolic tissues suggested TNF-α as a critical point that affected the co-development of inflammation and insulin resistance, but also pointed to the putatively protective roles of increased COX-2 and IL-17A signaling in the mediation of these pathophysiological states. Our results show that reduction of diet-induced inflammation confers partial protection against insulin resistance, but not obesity, and suggest the possibility of achieving overweight phenotypes that are accompanied by minimal insulin resistance if inflammation is controlled.
Collapse
Affiliation(s)
- Lyudmila Grodsky
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (L.G.); (M.W.); (T.R.)
- Department of Post-Baccalaureate Studies, University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte, NC 28223, USA
- School of Medicine, University of North Carolina at Chapel Hill, 150 Medical Drive, Chapel Hill, NC 27514, USA
| | - Mickey Wilson
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (L.G.); (M.W.); (T.R.)
| | - Thirumurugan Rathinasabapathy
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (L.G.); (M.W.); (T.R.)
| | - Slavko Komarnytsky
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (L.G.); (M.W.); (T.R.)
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University, 400 Dan Allen Drive, Raleigh, NC 27695, USA
| |
Collapse
|
12
|
Liu X, Yang J, Yan Y, Li Q, Huang RL. Unleashing the potential of adipose organoids: A revolutionary approach to combat obesity-related metabolic diseases. Theranostics 2024; 14:2075-2098. [PMID: 38505622 PMCID: PMC10945346 DOI: 10.7150/thno.93919] [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: 01/05/2024] [Accepted: 02/15/2024] [Indexed: 03/21/2024] Open
Abstract
Obesity-related metabolic diseases, including obesity, diabetes, hyperlipidemia, and non-alcoholic fatty liver diseases pose a significant threat to health. However, comprehensive pathogenesis exploration and effective therapy development are impeded by the limited availability of human models. Notably, advances in organoid technology enable the generation of adipose organoids that recapitulate structures and functions of native human adipose tissues to investigate mechanisms and develop corresponding treatments for obesity-related metabolic diseases. Here, we review the general principles, sources, and three-dimensional techniques for engineering adipose organoids, along with strategies to promote maturation. We also outline the application of white adipose organoids, primarily for disease modeling and drug screening, and highlight the therapeutic potential of thermogenic beige and brown adipose organoids in promoting weight loss and glucose and lipid metabolic homeostasis. We also discuss the challenges and prospects in the establishment and bench-to-bedside of adipose organoids, as well as their potential applications.
Collapse
Affiliation(s)
- Xingran Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Plastic and Reconstructive Surgery, Shanghai, China
| | - Jing Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Plastic and Reconstructive Surgery, Shanghai, China
| | - Yuxin Yan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Plastic and Reconstructive Surgery, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Plastic and Reconstructive Surgery, Shanghai, China
| | - Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Plastic and Reconstructive Surgery, Shanghai, China
| |
Collapse
|
13
|
Sorokin L, Corrêa LH. Whole-Mount Imaging of Adipose Tissue Macrophages. Methods Mol Biol 2024; 2713:307-322. [PMID: 37639132 DOI: 10.1007/978-1-0716-3437-0_21] [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: 08/29/2023]
Abstract
The adipose tissue comprises highly heterogeneous macrophage populations, which play critical roles in the regulation of adipose tissue function and dysfunction during health and disease. Whole-amount staining is a powerful technique for macrophage characterization within the 3D environment of the adipose tissue, enabling the visualization of different macrophage populations and their interaction with other cells within their in vivo niche. Due to the high-fat content and softness, freezing and sectioning of adipose tissue is difficult, and distortion of tissue morphology typically occurs, especially in the case of white adipose tissue. We describe here a whole-mount staining alternative for adipose tissue imaging that preserves all structures and allows high-resolution image acquisition. We address in a step-by-step manner how to perform immunofluorescence staining of different fat pads and how to optimally visualize cellular and acellular (extracellular matrix) constituents of the adipose tissue and its vasculature, as well as resident and infiltrating macrophage populations.
Collapse
Affiliation(s)
- Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, Cells-in-Motion Interfaculty Center (CIMIC), University of Münster, Münster, Germany
| | - Luis Henrique Corrêa
- Institute of Physiological Chemistry and Pathobiochemistry, Cells-in-Motion Interfaculty Center (CIMIC), University of Münster, Münster, Germany.
| |
Collapse
|
14
|
Raafat S, Ahmed R, Mowafi I, Adel H. Relation between the Epicardial Fat Thickness and the Cardiac Conduction System in Children and Adolescents with Diabetes. Horm Res Paediatr 2023; 97:496-508. [PMID: 38071957 PMCID: PMC11446301 DOI: 10.1159/000535630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 11/29/2023] [Indexed: 10/03/2024] Open
Abstract
INTRODUCTION Atherosclerosis in patients with type 1 diabetes starts early in childhood with subclinical abnormalities. The epicardial fat thickness (EFT) is a novel method for detecting these early changes. Furthermore, electrocardiographic markers may be altered in patients with diabetes owing to early cardiovascular changes. This study aimed to determine the relationship between EFT and electrocardiographic markers in children with type 1 diabetes mellitus. METHODS Children with type 1 diabetes who were followed up at the Alexandria University Children's Hospital Diabetes Clinic were enrolled in this study. The study recruited three groups of participants, including 20 patients with a diabetes duration of less than 5 years, 20 patients with a diabetes duration of 5 years or more, and 20 healthy controls. All participants were evaluated with emphasis on anthropometric measurements, fasting blood glucose levels, and lipid profile. HbA1c levels were measured in the cohort with diabetes. All participants underwent electrocardiography for measurement of P-wave dispersion, corrected QT interval and its dispersion, and Tp-e measurement. Echocardiography was performed to measure the EFT. RESULTS Among all participants, EFT was significantly higher in children with a diabetes duration of ≥5 years (p = 0.009). Furthermore, P-wave dispersion was significantly prolonged in children with diabetes compared to that in nondiabetics (p = 0.041). There was a statistically significant correlation between EFT and P-wave dispersion in patients with diabetes aged ≥5 years (p = 0.021). CONCLUSIONS Measurement of EFT by echocardiography is a novel and easy way to predict early cardiovascular changes in children with diabetes, including conduction system disorders.
Collapse
Affiliation(s)
- Shaymaa Raafat
- Pediatric Endocrinology and Diabetes Unit, Pediatric Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Reham Ahmed
- Pediatric Endocrinology and Diabetes Unit, Pediatric Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ihsan Mowafi
- Pediatric Endocrinology and Diabetes Unit, Pediatric Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Hani Adel
- Pediatric cardiology, Pediatric Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
15
|
Hetty S, Vranic M, Kamble PG, Lundqvist MH, Pereira MJ, Eriksson JW. CABLES1 expression is reduced in human subcutaneous adipose tissue in obesity and type 2 diabetes but may not directly impact adipocyte glucose and lipid metabolism. Adipocyte 2023; 12:2242997. [PMID: 37555665 PMCID: PMC10413912 DOI: 10.1080/21623945.2023.2242997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/16/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023] Open
Abstract
Cdk5 and Abl enzyme substrate 1 (CABLES1) is a cell cycle regulator that has previously been identified as a candidate gene for obesity-related phenotypes, but little is known about its role in adipose tissue metabolism. In this study, we explore the role of CABLES1 in obesity and type 2 diabetes (T2D) in human subcutaneous adipose tissue (SAT). We performed gene expression analysis of SAT obtained from subjects with and without T2D, and from a second validation cohort consisting of subjects without T2D. We used CRISPR/Cas9 genome editing to perform CABLES1 loss-of-function studies in human primary preadipocytes and assessed them functionally after differentiation. CABLES1 gene expression in SAT was decreased in T2D by almost 25%, and inversely associated with insulin resistance markers and hyperglycaemia. mRNA levels were reduced with increasing BMI and negatively correlated with obesity markers. We found that adipocytes are likely the main CABLES1-expressing cell type in SAT, but CABLES1 depletion in adipocytes caused no phenotypical changes in regards to differentiation, glucose uptake, or expression of key genes of adipocyte function. These findings suggest that CABLES1 gene expression in SAT might be altered in obesity and T2D as a consequence of metabolic dysregulation rather than being a causal factor.
Collapse
Affiliation(s)
- Susanne Hetty
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Milica Vranic
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Prasad G Kamble
- Innovation Strategies & External Liaison, Pharmaceutical Technologies & Development, AstraZeneca R&D, Mölndal, Sweden
| | - Martin H Lundqvist
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| |
Collapse
|
16
|
Brotman SM, Oravilahti A, Rosen JD, Alvarez M, Heinonen S, van der Kolk BW, Fernandes Silva L, Perrin HJ, Vadlamudi S, Pylant C, Deochand S, Basta PV, Valone JM, Narain MN, Stringham HM, Boehnke M, Kuusisto J, Love MI, Pietiläinen KH, Pajukanta P, Laakso M, Mohlke KL. Cell-Type Composition Affects Adipose Gene Expression Associations With Cardiometabolic Traits. Diabetes 2023; 72:1707-1718. [PMID: 37647564 PMCID: PMC10588284 DOI: 10.2337/db23-0365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023]
Abstract
Understanding differences in adipose gene expression between individuals with different levels of clinical traits may reveal the genes and mechanisms leading to cardiometabolic diseases. However, adipose is a heterogeneous tissue. To account for cell-type heterogeneity, we estimated cell-type proportions in 859 subcutaneous adipose tissue samples with bulk RNA sequencing (RNA-seq) using a reference single-nuclear RNA-seq data set. Cell-type proportions were associated with cardiometabolic traits; for example, higher macrophage and adipocyte proportions were associated with higher and lower BMI, respectively. We evaluated cell-type proportions and BMI as covariates in tests of association between >25,000 gene expression levels and 22 cardiometabolic traits. For >95% of genes, the optimal, or best-fit, models included BMI as a covariate, and for 79% of associations, the optimal models also included cell type. After adjusting for the optimal covariates, we identified 2,664 significant associations (P ≤ 2e-6) for 1,252 genes and 14 traits. Among genes proposed to affect cardiometabolic traits based on colocalized genome-wide association study and adipose expression quantitative trait locus signals, 25 showed a corresponding association between trait and gene expression levels. Overall, these results suggest the importance of modeling cell-type proportion when identifying gene expression associations with cardiometabolic traits. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Sarah M. Brotman
- Department of Genetics, The University of North Carolina, Chapel Hill, NC
| | - Anniina Oravilahti
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jonathan D. Rosen
- Department of Genetics, The University of North Carolina, Chapel Hill, NC
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Birgitta W. van der Kolk
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Hannah J. Perrin
- Department of Genetics, The University of North Carolina, Chapel Hill, NC
| | | | - Cortney Pylant
- Department of Epidemiology, The University of North Carolina, Chapel Hill, NC
| | - Sonia Deochand
- Department of Epidemiology, The University of North Carolina, Chapel Hill, NC
| | - Patricia V. Basta
- Department of Epidemiology, The University of North Carolina, Chapel Hill, NC
| | - Jordan M. Valone
- Department of Genetics, The University of North Carolina, Chapel Hill, NC
- UNC Neuroscience Center, The University of North Carolina, Chapel Hill, NC
| | - Morgan N. Narain
- Department of Genetics, The University of North Carolina, Chapel Hill, NC
- Curriculum of Toxicology and Environmental Medicine, The University of North Carolina, Chapel Hill, NC
| | - Heather M. Stringham
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Michael I. Love
- Department of Genetics, The University of North Carolina, Chapel Hill, NC
- Department of Biostatistics, The University of North Carolina, Chapel Hill, NC
| | - Kirsi H. Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
- Institute for Precision Health, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Karen L. Mohlke
- Department of Genetics, The University of North Carolina, Chapel Hill, NC
| |
Collapse
|
17
|
Moser J, Emous M, Heeringa P, Rodenhuis-Zybert IA. Mechanisms and pathophysiology of SARS-CoV-2 infection of the adipose tissue. Trends Endocrinol Metab 2023; 34:735-748. [PMID: 37673763 DOI: 10.1016/j.tem.2023.08.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 09/08/2023]
Abstract
Obesity is an independent risk factor for severe COVID-19, yet there remains a lack of consensus on the mechanisms underlying this relationship. A hypothesis that has garnered considerable attention suggests that SARS-CoV-2 disrupts adipose tissue function, either through direct infection or by indirect mechanisms. Indeed, recent reports have begun to shed some light on the important role that the adipose tissue plays during the acute phase of infection, as well as mediating long-term sequelae. In this review, we examine the evidence of extrapulmonary dissemination of SARS-CoV-2 to the adipose tissue. We discuss the mechanisms, acute and long-term implications, and possible management strategies to limit or ameliorate severe disease and long-term metabolic disturbances.
Collapse
Affiliation(s)
- Jill Moser
- Department of Critical Care, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Marloes Emous
- Center Obesity Northern Netherlands (CON), Department of Surgery, Medical Center Leeuwarden, Leeuwarden, the Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Izabela A Rodenhuis-Zybert
- Department of Medical Microbiology & Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
18
|
Mazitova AM, Márquez-Sánchez AC, Koltsova EK. Fat and inflammation: adipocyte-myeloid cell crosstalk in atherosclerosis. Front Immunol 2023; 14:1238664. [PMID: 37781401 PMCID: PMC10540690 DOI: 10.3389/fimmu.2023.1238664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Adipose tissue inflammation has been implicated in various chronic inflammatory diseases and cancer. Perivascular adipose tissue (PVAT) surrounds the aorta as an extra layer and was suggested to contribute to atherosclerosis development. PVAT regulates the function of endothelial and vascular smooth muscle cells in the aorta and represent a reservoir for various immune cells which may participate in aortic inflammation. Recent studies demonstrate that adipocytes also express various cytokine receptors and, therefore, may directly respond to inflammatory stimuli. Here we will summarize current knowledge on immune mechanisms regulating adipocyte activation and the crosstalk between myeloid cells and adipocytes in pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Aleksandra M. Mazitova
- Cedars-Sinai Cancer, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ana Cristina Márquez-Sánchez
- Cedars-Sinai Cancer, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ekaterina K. Koltsova
- Cedars-Sinai Cancer, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
19
|
Barroso LSS, Faria MHS, Souza-Gomes AF, Barros JLVM, Kakehasi AM, Vieira ELM, Simões E Silva AC, Nunes-Silva A. Acute and Chronic Effects of Strength Training on Plasma Levels of Adipokines in Man. Int J Sports Med 2023; 44:751-758. [PMID: 37429318 DOI: 10.1055/a-2079-1607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Adipose tissue is specialized cells that produce and release adipokines. Exercise may modulate adipokine production in adipocytes. The aim of this longitudinal study was to evaluate the acute and chronic effects of strength training (ST) on plasma levels of adiponectin, leptin, and resistin. Twelve untrained young male participants (23.42±2.67 years) were selected. The training protocol consisted of 3 exercises, with 3 sets of 65% of 1RM (one-repetition maximum) with pause of 90 s between sets with duration of 5 s/repetition (2 s conc/3 s ecc), 3 times a week for 10 weeks. Blood was collected at four time points: before and after the first ST session and before and after the last ST session. The comparisons between adipokine levels before and after the same training session showed acute changes, while the comparisons between levels before or after the first session versus before or after the last session revealed chronic alterations. ST increased adiponectin levels after the first exercise session in comparison to levels before this session [50 952 (46 568-51 894) pg/mL vs. 52 981 (49 901-54 467) pg/mL, p=0.019]. Similar differences were observed for resistin levels, which were higher after the last session compared to before [4 214.4 (±829) pg/mL vs. pre-S30 2 251.3 (±462.2) pg/mL, p=0.0008] and in the comparison between after the last and after the first ST sessions [4 214.4 (±829.0) pg/mL vs. 1 563.7 (±284.8) pg/mL, p=0.004]. Leptin levels acutely changed in the last training session. ST produced acute and chronic changes in plasma adipokines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Albená Nunes-Silva
- Physical Education School, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
20
|
Valenzuela PL, Carrera-Bastos P, Castillo-García A, Lieberman DE, Santos-Lozano A, Lucia A. Obesity and the risk of cardiometabolic diseases. Nat Rev Cardiol 2023; 20:475-494. [PMID: 36927772 DOI: 10.1038/s41569-023-00847-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 03/18/2023]
Abstract
The prevalence of obesity has reached pandemic proportions, and now approximately 25% of adults in Westernized countries have obesity. Recognized as a major health concern, obesity is associated with multiple comorbidities, particularly cardiometabolic disorders. In this Review, we present obesity as an evolutionarily novel condition, summarize the epidemiological evidence on its detrimental cardiometabolic consequences and discuss the major mechanisms involved in the association between obesity and the risk of cardiometabolic diseases. We also examine the role of potential moderators of this association, with evidence for and against the so-called 'metabolically healthy obesity phenotype', the 'fatness but fitness' paradox or the 'obesity paradox'. Although maintenance of optimal cardiometabolic status should be a primary goal in individuals with obesity, losing body weight and, particularly, excess visceral adiposity seems to be necessary to minimize the risk of cardiometabolic diseases.
Collapse
Affiliation(s)
- Pedro L Valenzuela
- Physical Activity and Health Research Group (PaHerg), Research Institute of Hospital 12 de Octubre ("i + 12"), Madrid, Spain.
- Department of Systems Biology, University of Alcalá, Alcalá de Henares, Spain.
| | - Pedro Carrera-Bastos
- Center for Primary Health Care Research, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain
| | | | - Daniel E Lieberman
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Alejandro Santos-Lozano
- Physical Activity and Health Research Group (PaHerg), Research Institute of Hospital 12 de Octubre ("i + 12"), Madrid, Spain
- Department of Health Sciences, European University Miguel de Cervantes, Valladolid, Spain
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain.
- CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain.
| |
Collapse
|
21
|
Morris I, Croes CA, Boes M, Kalkhoven E. Advanced omics techniques shed light on CD1d-mediated lipid antigen presentation to iNKT cells. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159292. [PMID: 36773690 DOI: 10.1016/j.bbalip.2023.159292] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/26/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Invariant natural killer T cells (iNKT cells) can be activated through binding antigenic lipid/CD1d complexes to their TCR. Antigenic lipids are processed, loaded, and displayed in complex with CD1d by lipid antigen presenting cells (LAPCs). The mechanism of lipid antigen presentation via CD1d is highly conserved with recent work showing adipocytes are LAPCs that, besides having a role in lipid storage, can activate iNKT cells and play an important role in systemic metabolic disease. Recent studies shed light on parameters potentially dictating cytokine output and how obesity-associated metabolic disease may affect such parameters. By following a lipid antigen's journey, we identify five key areas which may dictate cytokine skew: co-stimulation, structural properties of the lipid antigen, stability of lipid antigen/CD1d complexes, intracellular and extracellular pH, and intracellular and extracellular lipid environment. Recent publications indicate that the combination of advanced omics-type approaches and machine learning may be a fruitful way to interconnect these 5 areas, with the ultimate goal to provide new insights for therapeutic exploration.
Collapse
Affiliation(s)
- Imogen Morris
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584, CG, Utrecht, the Netherlands
| | - Cresci-Anne Croes
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, 6708WE Wageningen, the Netherlands
| | - Marianne Boes
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584, EA, Utrecht, the Netherlands; Department of Paediatric Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584, EA, Utrecht, the Netherlands
| | - Eric Kalkhoven
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584, CG, Utrecht, the Netherlands.
| |
Collapse
|
22
|
Baez AS, Ortiz-Whittingham LR, Tarfa H, Osei Baah F, Thompson K, Baumer Y, Powell-Wiley TM. Social determinants of health, health disparities, and adiposity. Prog Cardiovasc Dis 2023; 78:17-26. [PMID: 37178992 PMCID: PMC10330861 DOI: 10.1016/j.pcad.2023.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023]
Abstract
Social determinants of health (SDoH), or the socioeconomic, environmental, and psychosocial conditions in which individuals spend their daily lives, substantially influence obesity as a cardiovascular disease (CVD) risk factor. The coronavirus disease 2019 (COVID-19) pandemic highlighted the converging epidemics of obesity, CVD, and social inequities globally. Obesity and CVD serve as independent risk factors for COVID-19 severity and lower-resourced populations most impacted by adverse SDoH have the highest COVID-19 mortality rates. Better understanding the interplay between social and biologic factors that contribute to obesity-related CVD disparities are important to equitably address obesity across populations. Despite efforts to investigate SDoH and their biologic effects as drivers of health disparities, the connections between SDoH and obesity remain incompletely understood. This review aims to highlight the relationships between socioeconomic, environmental, and psychosocial factors and obesity. We also present potential biologic factors that may play a role in the biology of adversity, or link SDoH to adiposity and poor adipo-cardiology outcomes. Finally, we provide evidence for multi-level obesity interventions targeting multiple aspects of SDoH. Throughout, we emphasize areas for future research to tailor health equity-promoting interventions across populations to reduce obesity and obesity-related CVD disparities.
Collapse
Affiliation(s)
- Andrew S Baez
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg 10-CRC, 5-5330, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Lola R Ortiz-Whittingham
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg 10-CRC, 5-5330, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Hannatu Tarfa
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg 10-CRC, 5-5330, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Foster Osei Baah
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg 10-CRC, 5-5330, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Keitra Thompson
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg 10-CRC, 5-5330, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg 10-CRC, 5-5330, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Tiffany M Powell-Wiley
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg 10-CRC, 5-5330, 10 Center Drive, Bethesda, MD 20892, USA; Intramural Research Program, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
23
|
Badran S, Doi SA, Hamdi M, Hammouda A, Alharami S, Clark J, H Musa OA, Abou-Samra AB, M Habib A. Metabolic aspects of surgical subcutaneous fat removal: An umbrella review and implications for future research. BIOMOLECULES & BIOMEDICINE 2023; 23:235-247. [PMID: 36200436 PMCID: PMC10113936 DOI: 10.17305/bjbms.2022.8175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022]
Abstract
Although obesity is a preventable disease, maintaining a normal body weight can be very challenging and difficult, which has led to a significant increase in the demand for surgical subcutaneous fat removal (SSFR) to improve physical appearance. The need for SSFR is further exacerbated because of the global rise in the number of bariatric surgeries, which is currently the single most durable intervention for mitigating obesity. Fat tissue is now recognized as a vital endocrine organ that produces several bioactive proteins. Thus, SSFR-mediated weight (fat) loss can potentially have significant metabolic effects; however, currently, there is no consensus on this issue. This review focuses on the metabolic sequelae after SSFR interventions for dealing with cosmetic body appearance. Data was extracted from existing systematic reviews and the diversity of possible metabolic changes after SSFR are reported along with gaps in the knowledge and future directions for research and practice. We conclude that there is a potential for metabolic sequelae after SSFR interventions and their clinical implications for the safety of the procedures as well as for our understanding of subcutaneous adipose tissue biology and insulin resistance are discussed.
Collapse
Affiliation(s)
- Saif Badran
- Department of Population Medicine, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Suhail A Doi
- Department of Population Medicine, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Moustapha Hamdi
- Department of Plastic and Reconstructive Surgery, Brussels University Hospital, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Atalla Hammouda
- Department of Plastic Surgery, Hamad General Hospital, Doha, Qatar
| | - Sara Alharami
- Department of Plastic Surgery, Hamad General Hospital, Doha, Qatar
| | - Justin Clark
- Institute for Evidence-Based Healthcare, Faculty of Health Sciences & Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Omran A H Musa
- Department of Population Medicine, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Abdul-Badi Abou-Samra
- Department of Medicine, Weill Cornell Medicine Qatar, Qatar Foundation, Doha, Qatar
- Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
24
|
An integrated single cell and spatial transcriptomic map of human white adipose tissue. Nat Commun 2023; 14:1438. [PMID: 36922516 PMCID: PMC10017705 DOI: 10.1038/s41467-023-36983-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
To date, single-cell studies of human white adipose tissue (WAT) have been based on small cohort sizes and no cellular consensus nomenclature exists. Herein, we performed a comprehensive meta-analysis of publicly available and newly generated single-cell, single-nucleus, and spatial transcriptomic results from human subcutaneous, omental, and perivascular WAT. Our high-resolution map is built on data from ten studies and allowed us to robustly identify >60 subpopulations of adipocytes, fibroblast and adipogenic progenitors, vascular, and immune cells. Using these results, we deconvolved spatial and bulk transcriptomic data from nine additional cohorts to provide spatial and clinical dimensions to the map. This identified cell-cell interactions as well as relationships between specific cell subtypes and insulin resistance, dyslipidemia, adipocyte volume, and lipolysis upon long-term weight changes. Altogether, our meta-map provides a rich resource defining the cellular and microarchitectural landscape of human WAT and describes the associations between specific cell types and metabolic states.
Collapse
|
25
|
Dou J, Thangaraj SV, Puttabyatappa M, Elangovan VR, Bakulski K, Padmanabhan V. Developmental programming: Adipose depot-specific regulation of non-coding RNAs and their relation to coding RNA expression in prenatal testosterone and prenatal bisphenol-A -treated female sheep. Mol Cell Endocrinol 2023; 564:111868. [PMID: 36708980 PMCID: PMC10069610 DOI: 10.1016/j.mce.2023.111868] [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: 12/14/2022] [Revised: 01/19/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
Inappropriate developmental exposure to steroids is linked to metabolic disorders. Prenatal testosterone excess or bisphenol A (BPA, an environmental estrogen mimic) leads to insulin resistance and adipocyte disruptions in female lambs. Adipocytes are key regulators of insulin sensitivity. Metabolic tissue-specific differences in insulin sensitivity coupled with adipose depot-specific changes in key mRNAs, were previously observed with prenatal steroid exposure. We hypothesized that depot-specific changes in the non-coding RNA (ncRNA) - regulators of gene expression would account for the direction of changes seen in mRNAs. Non-coding RNA (lncRNA, miRNA, snoRNA, snRNA) from various adipose depots of prenatal testosterone and BPA-treated animals were sequenced. Adipose depot-specific changes in the ncRNA that are consistent with the depot-specific mRNA expression in terms of directionality of changes and functional implications in insulin resistance, adipocyte differentiation and cardiac hypertrophy were found. Importantly, the adipose depot-specific ncRNA changes were model-specific and mutually exclusive, suggestive of different regulatory entry points in this regulation.
Collapse
Affiliation(s)
- John Dou
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | - Kelly Bakulski
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | | |
Collapse
|
26
|
Bogard G, Barthelemy J, Hantute-Ghesquier A, Sencio V, Brito-Rodrigues P, Séron K, Robil C, Flourens A, Pinet F, Eberlé D, Trottein F, Duterque-Coquillaud M, Wolowczuk I. SARS-CoV-2 infection induces persistent adipose tissue damage in aged golden Syrian hamsters. Cell Death Dis 2023; 14:75. [PMID: 36725844 PMCID: PMC9891765 DOI: 10.1038/s41419-023-05574-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 02/03/2023]
Abstract
Coronavirus disease 2019 (COVID-19, caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2)) is primarily a respiratory illness. However, various extrapulmonary manifestations have been reported in patients with severe forms of COVID-19. Notably, SARS-CoV-2 was shown to directly trigger white adipose tissue (WAT) dysfunction, which in turn drives insulin resistance, dyslipidemia, and other adverse outcomes in patients with COVID-19. Although advanced age is the greatest risk factor for COVID-19 severity, published data on the impact of SARS-CoV-2 infection on WAT in aged individuals are scarce. Here, we characterized the response of subcutaneous and visceral WAT depots to SARS-CoV-2 infection in young adult and aged golden hamsters. In both age groups, infection was associated with a decrease in adipocyte size in the two WAT depots; this effect was partly due to changes in tissue's lipid metabolism and persisted for longer in aged hamsters than in young-adult hamsters. In contrast, only the subcutaneous WAT depot contained crown-like structures (CLSs) in which dead adipocytes were surrounded by SARS-CoV-2-infected macrophages, some of them forming syncytial multinucleated cells. Importantly, older age predisposed to a unique manifestation of viral disease in the subcutaneous WAT depot during SARS-CoV-2 infection; the persistence of very large CLSs was indicative of an age-associated defect in the clearance of dead adipocytes by macrophages. Moreover, we uncovered age-related differences in plasma lipid profiles during SARS-CoV-2 infection. These data suggest that the WAT's abnormal response to SARS-CoV-2 infection may contribute to the greater severity of COVID-19 observed in elderly patients.
Collapse
Affiliation(s)
- Gemma Bogard
- Univ. Lille, Institut National de la Santé et de la Recherche Médicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019-UMR9017-Center for Infection and Immunity of Lille (CIIL), F-59000, Lille, France
| | - Johanna Barthelemy
- Univ. Lille, Institut National de la Santé et de la Recherche Médicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019-UMR9017-Center for Infection and Immunity of Lille (CIIL), F-59000, Lille, France
| | - Aline Hantute-Ghesquier
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - Valentin Sencio
- Univ. Lille, Institut National de la Santé et de la Recherche Médicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019-UMR9017-Center for Infection and Immunity of Lille (CIIL), F-59000, Lille, France
| | - Patricia Brito-Rodrigues
- Univ. Lille, Institut National de la Santé et de la Recherche Médicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019-UMR9017-Center for Infection and Immunity of Lille (CIIL), F-59000, Lille, France
| | - Karin Séron
- Univ. Lille, Institut National de la Santé et de la Recherche Médicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019-UMR9017-Center for Infection and Immunity of Lille (CIIL), F-59000, Lille, France
| | - Cyril Robil
- Univ. Lille, Institut National de la Santé et de la Recherche Médicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019-UMR9017-Center for Infection and Immunity of Lille (CIIL), F-59000, Lille, France
| | - Anne Flourens
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - Florence Pinet
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Delphine Eberlé
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000, Lille, France
| | - François Trottein
- Univ. Lille, Institut National de la Santé et de la Recherche Médicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019-UMR9017-Center for Infection and Immunity of Lille (CIIL), F-59000, Lille, France
| | - Martine Duterque-Coquillaud
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - Isabelle Wolowczuk
- Univ. Lille, Institut National de la Santé et de la Recherche Médicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019-UMR9017-Center for Infection and Immunity of Lille (CIIL), F-59000, Lille, France.
| |
Collapse
|
27
|
Burkhardt LM, Bucher CH, Löffler J, Rinne C, Duda GN, Geissler S, Schulz TJ, Schmidt-Bleek K. The benefits of adipocyte metabolism in bone health and regeneration. Front Cell Dev Biol 2023; 11:1104709. [PMID: 36895792 PMCID: PMC9988968 DOI: 10.3389/fcell.2023.1104709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Patients suffering from musculoskeletal diseases must cope with a diminished quality of life and an increased burden on medical expenses. The interaction of immune cells and mesenchymal stromal cells during bone regeneration is one of the key requirements for the restoration of skeletal integrity. While stromal cells of the osteo-chondral lineage support bone regeneration, an excessive accumulation of cells of the adipogenic lineage is thought to promote low-grade inflammation and impair bone regeneration. Increasing evidence indicates that pro-inflammatory signaling from adipocytes is responsible for various chronic musculoskeletal diseases. This review aims to summarize the features of bone marrow adipocytes by phenotype, function, secretory features, metabolic properties and their impact on bone formation. In detail, the master regulator of adipogenesis and prominent diabetes drug target, peroxisome proliferator-activated receptor γ (PPARG), will be debated as a potential therapeutic approach to enhance bone regeneration. We will explore the possibilities of using clinically established PPARG agonists, the thiazolidinediones (TZDs), as a treatment strategy to guide the induction of a pro-regenerative, metabolically active bone marrow adipose tissue. The impact of this PPARG induced bone marrow adipose tissue type on providing the necessary metabolites to sustain osteogenic-as well as beneficial immune cells during bone fracture healing will be highlighted.
Collapse
Affiliation(s)
- Lisa-Marie Burkhardt
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Christian H Bucher
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Julia Löffler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Charlotte Rinne
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Sven Geissler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
28
|
Khudyakov JI, Allen KN, Crocker DE, Trost NS, Roberts AH, Pirard L, Debier C, Piotrowski ER, Vázquez-Medina JP. Comprehensive molecular and morphological resolution of blubber stratification in a deep-diving, fasting-adapted seal. Front Physiol 2022; 13:1057721. [PMID: 36589428 PMCID: PMC9795062 DOI: 10.3389/fphys.2022.1057721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Blubber is a modified subcutaneous adipose tissue in marine mammals that provides energy storage, thermoregulation, hydrodynamic locomotion, and buoyancy. Blubber displays vertical stratification by lipid content, fatty acid composition, and vascularization, leading to the assumption that deeper blubber layers are metabolically active, while superficial layers are mainly structural and thermoregulatory. However, few studies have examined functional stratification of marine mammal blubber directly, especially in pinnipeds. We characterized morphological and transcriptional differences across blubber layers in the northern elephant seal, a deep-diving and fasting-adapted phocid. We collected blubber from seals early in their fasting period and divided blubber cores into three similarly sized portions. We hypothesized that the innermost blubber portion would have higher 1) heterogeneity in adipocyte size, 2) microvascular density, and 3) expression of genes associated with metabolism and hormone signaling than outer blubber. We found that adipocyte area and variance increased from outermost (skin-adjacent) to innermost (muscle-adjacent) blubber layers, suggesting that inner blubber has a higher capacity for lipid storage and turnover than outer blubber. Inner blubber had a higher proportion of CD144+ endothelial cells, suggesting higher microvascular density. In contrast, outer blubber had a higher proportion of CD4+ immune cells than inner blubber, suggesting higher capacity for response to tissue injury. Transcriptome analysis identified 61 genes that were differentially expressed between inner and outer blubber layers, many of which have not been studied previously in marine mammals. Based on known functions of these genes in other mammals, we suggest that inner blubber has potentially higher 1) adipogenic capacity, 2) cellular diversity, and 3) metabolic and neuroendocrine signaling activity, while outer blubber may have higher 1) extracellular matrix synthesis activity and 2) responsiveness to pathogens and cell stressors. We further characterized expression of nine genes of interest identified by transcriptomics and two adipokines with higher precision across blubber layers using targeted assays. Our study provides functional insights into stratification of blubber in marine mammals and a molecular key, including CD144, CD4, HMGCS2, GABRG2, HCAR2, and COL1A2, for distinguishing blubber layers for physiological and functional studies in seals.
Collapse
Affiliation(s)
- J. I. Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States,*Correspondence: J. I. Khudyakov,
| | - K. N. Allen
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, United States
| | - D. E. Crocker
- Department of Biology, Sonoma State University, Rohnert Park, CA, United States
| | - N. S. Trost
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | - A. H. Roberts
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | - L. Pirard
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la Neuve, Belgium
| | - C. Debier
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la Neuve, Belgium
| | - E. R. Piotrowski
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | - J. P. Vázquez-Medina
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
29
|
Hanusek K, Karczmarski J, Litwiniuk A, Urbańska K, Ambrozkiewicz F, Kwiatkowski A, Martyńska L, Domańska A, Bik W, Paziewska A. Obesity as a Risk Factor for Breast Cancer-The Role of miRNA. Int J Mol Sci 2022; 23:ijms232415683. [PMID: 36555323 PMCID: PMC9779381 DOI: 10.3390/ijms232415683] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Breast cancer (BC) is the most common cancer diagnosed among women in the world, with an ever-increasing incidence rate. Due to the dynamic increase in the occurrence of risk factors, including obesity and related metabolic disorders, the search for new regulatory mechanisms is necessary. This will help a complete understanding of the pathogenesis of breast cancer. The review presents the mechanisms of obesity as a factor that increases the risk of developing breast cancer and that even initiates the cancer process in the female population. The mechanisms presented in the paper relate to the inflammatory process resulting from current or progressive obesity leading to cell metabolism disorders and disturbed hormonal metabolism. All these processes are widely regulated by the action of microRNAs (miRNAs), which may constitute potential biomarkers influencing the pathogenesis of breast cancer and may be a promising target of anti-cancer therapies.
Collapse
Affiliation(s)
- Karolina Hanusek
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Jakub Karczmarski
- Department of Neuroendocrinology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Anna Litwiniuk
- Department of Neuroendocrinology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Katarzyna Urbańska
- Department of General, Oncological, Metabolic and Thoracic Surgery, Military Institute of Medicine, 128 Szaserów St, 04-141 Warsaw, Poland
| | - Filip Ambrozkiewicz
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1665/76, 32300 Pilsen, Czech Republic
| | - Andrzej Kwiatkowski
- Department of General, Oncological, Metabolic and Thoracic Surgery, Military Institute of Medicine, 128 Szaserów St, 04-141 Warsaw, Poland
| | - Lidia Martyńska
- Department of Neuroendocrinology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Anita Domańska
- Department of Neuroendocrinology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Wojciech Bik
- Department of Neuroendocrinology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Agnieszka Paziewska
- Department of Neuroendocrinology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
- Faculty of Medical and Health Sciences, Institute of Health Sciences, Siedlce University of Natural Sciences and Humanities, 08-110 Siedlce, Poland
- Correspondence:
| |
Collapse
|
30
|
Scamfer SR, Lee MD, Hilgendorf KI. Ciliary control of adipocyte progenitor cell fate regulates energy storage. Front Cell Dev Biol 2022; 10:1083372. [PMID: 36561368 PMCID: PMC9763467 DOI: 10.3389/fcell.2022.1083372] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
The primary cilium is a cellular sensory organelle found in most cells in our body. This includes adipocyte progenitor cells in our adipose tissue, a complex organ involved in energy storage, endocrine signaling, and thermogenesis. Numerous studies have shown that the primary cilium plays a critical role in directing the cell fate of adipocyte progenitor cells in multiple adipose tissue types. Accordingly, diseases with dysfunctional cilia called ciliopathies have a broad range of clinical manifestations, including obesity and diabetes. This review summarizes our current understanding of how the primary cilium regulates adipocyte progenitor cell fate in multiple contexts and illustrates the importance of the primary cilium in regulating energy storage and adipose tissue function.
Collapse
Affiliation(s)
| | | | - Keren I. Hilgendorf
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
31
|
Poltronieri TS, Pérsico RS, Falcetta FS, Viana LV. Changes in Body Adiposity in Women Undergoing Breast Cancer Treatment: A Scoping Review. Nutr Cancer 2022; 74:3431-3445. [DOI: 10.1080/01635581.2022.2081341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Taiara S. Poltronieri
- Graduate Program in Medical Sciences, Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Hospital de Clinicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Raquel S. Pérsico
- Graduate Program in Medical Sciences, Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Hospital de Clinicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Frederico S. Falcetta
- Graduate Program in Medical Sciences, Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Hospital de Clinicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Luciana V. Viana
- Graduate Program in Medical Sciences, Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Hospital de Clinicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
32
|
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: 7.0] [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
|
33
|
Czaja-Stolc S, Potrykus M, Stankiewicz M, Kaska Ł, Małgorzewicz S. Pro-Inflammatory Profile of Adipokines in Obesity Contributes to Pathogenesis, Nutritional Disorders, and Cardiovascular Risk in Chronic Kidney Disease. Nutrients 2022; 14:nu14071457. [PMID: 35406070 PMCID: PMC9002635 DOI: 10.3390/nu14071457] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 01/27/2023] Open
Abstract
Obesity is a disease which leads to the development of many other disorders. Excessive accumulation of lipids in adipose tissue (AT) leads to metabolic changes, including hypertrophy of adipocytes, macrophage migration, changes in the composition of immune cells, and impaired secretion of adipokines. Adipokines are cytokines produced by AT and greatly influence human health. Obesity and the pro-inflammatory profile of adipokines lead to the development of chronic kidney disease (CKD) through different mechanisms. In obesity and adipokine profile, there are gender differences that characterize the male gender as more susceptible to metabolic disorders accompanying obesity, including impaired renal function. The relationship between impaired adipokine secretion and renal disease is two-sided. In the developed CKD, the concentration of adipokines in the serum is additionally disturbed due to their insufficient excretion by the excretory system caused by renal pathology. Increased levels of adipokines affect the nutritional status and cardiovascular risk (CVR) of patients with CKD. This article aims to systematize the current knowledge on the influence of obesity, AT, and adipokine secretion disorders on the pathogenesis of CKD and their influence on nutritional status and CVR in patients with CKD.
Collapse
Affiliation(s)
- Sylwia Czaja-Stolc
- Department of Clinical Nutrition, Medical University of Gdansk, 80-211 Gdańsk, Poland; (M.S.); (S.M.)
- Correspondence: ; Tel.: +48-(58)-349-27-24
| | - Marta Potrykus
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, 80-211 Gdańsk, Poland; (M.P.); (Ł.K.)
| | - Marta Stankiewicz
- Department of Clinical Nutrition, Medical University of Gdansk, 80-211 Gdańsk, Poland; (M.S.); (S.M.)
| | - Łukasz Kaska
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, 80-211 Gdańsk, Poland; (M.P.); (Ł.K.)
| | - Sylwia Małgorzewicz
- Department of Clinical Nutrition, Medical University of Gdansk, 80-211 Gdańsk, Poland; (M.S.); (S.M.)
| |
Collapse
|
34
|
Tan K, Naylor MJ. The Influence of Modifiable Factors on Breast and Prostate Cancer Risk and Disease Progression. Front Physiol 2022; 13:840826. [PMID: 35330933 PMCID: PMC8940211 DOI: 10.3389/fphys.2022.840826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/11/2022] [Indexed: 12/31/2022] Open
Abstract
Breast and prostate cancers are among the most commonly diagnosed cancers worldwide, and together represented almost 20% of all new cancer diagnoses in 2020. For both cancers, the primary treatment options are surgical resection and sex hormone deprivation therapy, highlighting the initial dependence of these malignancies on the activity of both endogenous and exogenous hormones. Cancer cell phenotype and patient prognosis is not only determined by the collection of specific gene mutations, but through the interaction and influence of a wide range of different local and systemic components. While genetic risk factors that contribute to the development of these cancers are well understood, increasing epidemiological evidence link modifiable lifestyle factors such as physical exercise, diet and weight management, to drivers of disease progression such as inflammation, transcriptional activity, and altered biochemical signaling pathways. As a result of this significant impact, it is estimated that up to 50% of cancer cases in developed countries could be prevented with changes to lifestyle and environmental factors. While epidemiological studies of modifiable risk factors and research of the biological mechanisms exist mostly independently, this review will discuss how advances in our understanding of the metabolic, protein and transcriptional pathways altered by modifiable lifestyle factors impact cancer cell physiology to influence breast and prostate cancer risk and prognosis.
Collapse
Affiliation(s)
| | - Matthew J. Naylor
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
35
|
The Shades of Grey in Adipose Tissue Reprogramming. Biosci Rep 2022; 42:230844. [PMID: 35211733 PMCID: PMC8905306 DOI: 10.1042/bsr20212358] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/22/2022] Open
Abstract
The adipose tissue (AT) has a major role in contributing to obesity-related pathologies through regulating systemic immunometabolism. The pathogenicity of the AT is underpinned by its remarkable plasticity to be reprogrammed during obesity, in the perspectives of tissue morphology, extracellular matrix (ECM) composition, angiogenesis, immunometabolic homoeostasis and circadian rhythmicity. Dysregulation in these features escalates the pathogenesis conferred by this endometabolic organ. Intriguingly, the potential to be reprogrammed appears to be an Achilles’ heel of the obese AT that can be targeted for the management of obesity and its associated comorbidities. Here, we provide an overview of the reprogramming processes of white AT (WAT), with a focus on their dynamics and pleiotropic actions over local and systemic homoeostases, followed by a discussion of potential strategies favouring therapeutic reprogramming. The potential involvement of AT remodelling in the pathogenesis of COVID-19 is also discussed.
Collapse
|
36
|
Affiliation(s)
- Aaron M Cypess
- From the Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
37
|
Exercise Inhibits NLRP3 Inflammasome Activation in Obese Mice via the Anti-Inflammatory Effect of Meteorin-like. Cells 2021; 10:cells10123480. [PMID: 34943988 PMCID: PMC8700724 DOI: 10.3390/cells10123480] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/19/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Obesity is associated with chronic low-grade inflammation. The benefits of exercise are partly attributed to its anti-inflammatory effect, but whether exercise can regulate NLRP3 inflammasome activation in obese adipose tissue remains unknown. Meteorin-like (METRNL), a recently discovered myokine, has been implicated in mediating the effect of exercise on metabolism. Herein, we examined the effect of exercise and METRNL on NLRP3 inflammasome activation. High-fat diet (HFD)-induced obese mice were subjected to treadmill exercise for 8 weeks. A subgroup of HFD mice was switched to normal chow with the exercise intervention. Exercise and diet attenuated weight gain, fat accumulation, and insulin resistance in obese mice. In addition, exercise downregulated gene and protein levels of inflammasome markers, including NLRP3 and caspase-1, in adipose tissue. In isolated bone marrow-derived macrophages, activation of NLRP3 inflammasome was suppressed in the exercise group, as confirmed by the downregulation of IL-1β and IL-18. Exercise significantly enhanced the expression of METRNL in various muscle depots, and further in vitro analysis revealed that recombinant METRNL treatment inhibited IL-1β secretion in macrophages. In conclusion, exercise exerts its anti-inflammatory action by suppressing adipose tissue NLRP3 inflammasome, and this is, in part, associated with METRNL induction in muscle and its anti-inflammatory effects in macrophages.
Collapse
|
38
|
Adipose Tissue Dysfunctions in Response to an Obesogenic Diet Are Reduced in Mice after Transgenerational Supplementation with Omega 3 Fatty Acids. Metabolites 2021; 11:metabo11120838. [PMID: 34940596 PMCID: PMC8706165 DOI: 10.3390/metabo11120838] [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: 11/03/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 11/23/2022] Open
Abstract
Obesity is characterized by profound alterations in adipose tissue (AT) biology, leading to whole body metabolic disturbances such as insulin resistance and cardiovascular diseases. These alterations are related to the development of a local inflammation, fibrosis, hypertrophy of adipocytes, and dysregulation in energy homeostasis, notably in visceral adipose tissue (VAT). Omega 3 (n-3) fatty acids (FA) have been described to possess beneficial effects against obesity-related disorders, including in the AT; however, the long-term effect across generations remains unknown. The current study was conducted to identify if supplementation with n-3 polyunsaturated FA (PUFA) for three generations could protect from the consequences of an obesogenic diet in VAT. Young mice from the third generation of a lineage receiving a daily supplementation (1% of the diet) with fish oil rich in eicosapentaenoic acid (EPA) or an isocaloric amount of sunflower oil, were fed a high-fat, high-sugar content diet for 4 months. We explore the transcriptomic adaptations in each lineage using DNA microarray in VAT and bioinformatic exploration of biological regulations using online databases. Transgenerational intake of EPA led to a reduced activation of inflammatory processes, perturbation in metabolic homeostasis, cholesterol metabolism, and mitochondrial functions in response to the obesogenic diet as compared to control mice from a control lineage. This suggests that the continuous intake of long chain n-3 PUFA could be preventive in situations of oversupply of energy-dense, nutrient-poor foods.
Collapse
|
39
|
Perrin HJ, Currin KW, Vadlamudi S, Pandey GK, Ng KK, Wabitsch M, Laakso M, Love MI, Mohlke KL. Chromatin accessibility and gene expression during adipocyte differentiation identify context-dependent effects at cardiometabolic GWAS loci. PLoS Genet 2021; 17:e1009865. [PMID: 34699533 PMCID: PMC8570510 DOI: 10.1371/journal.pgen.1009865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/05/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022] Open
Abstract
Chromatin accessibility and gene expression in relevant cell contexts can guide identification of regulatory elements and mechanisms at genome-wide association study (GWAS) loci. To identify regulatory elements that display differential activity across adipocyte differentiation, we performed ATAC-seq and RNA-seq in a human cell model of preadipocytes and adipocytes at days 4 and 14 of differentiation. For comparison, we created a consensus map of ATAC-seq peaks in 11 human subcutaneous adipose tissue samples. We identified 58,387 context-dependent chromatin accessibility peaks and 3,090 context-dependent genes between all timepoint comparisons (log2 fold change>1, FDR<5%) with 15,919 adipocyte- and 18,244 preadipocyte-dependent peaks. Adipocyte-dependent peaks showed increased overlap (60.1%) with Roadmap Epigenomics adipocyte nuclei enhancers compared to preadipocyte-dependent peaks (11.5%). We linked context-dependent peaks to genes based on adipocyte promoter capture Hi-C data, overlap with adipose eQTL variants, and context-dependent gene expression. Of 16,167 context-dependent peaks linked to a gene, 5,145 were linked by two or more strategies to 1,670 genes. Among GWAS loci for cardiometabolic traits, adipocyte-dependent peaks, but not preadipocyte-dependent peaks, showed significant enrichment (LD score regression P<0.005) for waist-to-hip ratio and modest enrichment (P < 0.05) for HDL-cholesterol. We identified 659 peaks linked to 503 genes by two or more approaches and overlapping a GWAS signal, suggesting a regulatory mechanism at these loci. To identify variants that may alter chromatin accessibility between timepoints, we identified 582 variants in 454 context-dependent peaks that demonstrated allelic imbalance in accessibility (FDR<5%), of which 55 peaks also overlapped GWAS variants. At one GWAS locus for palmitoleic acid, rs603424 was located in an adipocyte-dependent peak linked to SCD and exhibited allelic differences in transcriptional activity in adipocytes (P = 0.003) but not preadipocytes (P = 0.09). These results demonstrate that context-dependent peaks and genes can guide discovery of regulatory variants at GWAS loci and aid identification of regulatory mechanisms. Cardiovascular and metabolic diseases are widespread, and an increased understanding of genetic mechanisms behind these diseases could improve treatment. Chromatin accessibility and gene expression in relevant cell contexts can guide identification of regulatory elements and genetic mechanisms for disease traits. A relevant context for cardiovascular and metabolic disease traits is adipocyte differentiation. To identify regulatory elements and genes that display differences in activity during adipocyte differentiation, we profiled chromatin accessibility and gene expression in a human cell model of preadipocytes and adipocytes. We identified chromatin regions that change accessibility during differentiation and predicted genes they may affect. We also linked these chromatin regions to genetic variants associated with risk of disease. At one genomic region linked to fatty acids, a chromatin region more accessible in adipocytes linked to a fatty acid synthesis gene and exhibited allelic differences in transcriptional activity in adipocytes but not preadipocytes. These results demonstrate that chromatin regions and genes that change during cell context can guide discovery of regulatory variants and aid identification of disease mechanisms.
Collapse
Affiliation(s)
- Hannah J. Perrin
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Kevin W. Currin
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Swarooparani Vadlamudi
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Gautam K. Pandey
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Kenneth K. Ng
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Martin Wabitsch
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Michael I. Love
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
40
|
Modulation of Cardiac Arrhythmogenesis by Epicardial Adipose Tissue: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 78:1730-1745. [PMID: 34674819 DOI: 10.1016/j.jacc.2021.08.037] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 08/06/2021] [Accepted: 08/25/2021] [Indexed: 01/04/2023]
Abstract
Obesity is a significant risk factor for arrhythmic cardiovascular death. Interactions between epicardial adipose tissue (EAT) and myocytes are thought to play a key role in the development of arrhythmias. In this review, the authors investigate the influence of EAT on arrhythmogenesis. First, they summarize electrocardiographic evidence showing the association between increased EAT volume and atrial and ventricular conduction delay. Second, they detail the structural cross talk between EAT and the heart and its arrhythmogenicity. Adipose tissue infiltration within the myocardium constitutes an anatomical obstacle to cardiac excitation. It causes activation delay and increases the risk of arrhythmias. Intercellular electrical coupling between cardiomyocytes and EAT can further slow conduction and increase the risk of block, favoring re-entry and arrhythmias. Finally, EAT secretes multiple substances that influence cardiomyocyte electrophysiology either by modulating ion currents and electrical coupling or by stimulating fibrosis. Thus, structural and paracrine cross talk between EAT and cardiomyocytes facilitates arrhythmias.
Collapse
|
41
|
Kolnes KJ, Petersen MH, Lien-Iversen T, Højlund K, Jensen J. Effect of Exercise Training on Fat Loss-Energetic Perspectives and the Role of Improved Adipose Tissue Function and Body Fat Distribution. Front Physiol 2021; 12:737709. [PMID: 34630157 PMCID: PMC8497689 DOI: 10.3389/fphys.2021.737709] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022] Open
Abstract
In obesity, excessive abdominal fat, especially the accumulation of visceral adipose tissue (VAT), increases the risk of metabolic disorders, such as type 2 diabetes mellitus (T2DM), cardiovascular disease, and non-alcoholic fatty liver disease. Excessive abdominal fat is associated with adipose tissue dysfunction, leading to systemic low-grade inflammation, fat overflow, ectopic lipid deposition, and reduced insulin sensitivity. Physical activity is recommended for primary prevention and treatment of obesity, T2DM, and related disorders. Achieving a stable reduction in body weight with exercise training alone has not shown promising effects on a population level. Because fat has a high energy content, a large amount of exercise training is required to achieve weight loss. However, even when there is no weight loss, exercise training is an effective method of improving body composition (increased muscle mass and reduced fat) as well as increasing insulin sensitivity and cardiorespiratory fitness. Compared with traditional low-to-moderate-intensity continuous endurance training, high-intensity interval training (HIIT) and sprint interval training (SIT) are more time-efficient as exercise regimens and produce comparable results in reducing total fat mass, as well as improving cardiorespiratory fitness and insulin sensitivity. During high-intensity exercise, carbohydrates are the main source of energy, whereas, with low-intensity exercise, fat becomes the predominant energy source. These observations imply that HIIT and SIT can reduce fat mass during bouts of exercise despite being associated with lower levels of fat oxidation. In this review, we explore the effects of different types of exercise training on energy expenditure and substrate oxidation during physical activity, and discuss the potential effects of exercise training on adipose tissue function and body fat distribution.
Collapse
Affiliation(s)
| | | | - Teodor Lien-Iversen
- Department of Internal Medicine, Randers Regional Hospital, Randers, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Jørgen Jensen
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| |
Collapse
|
42
|
Baritussio A, Williams MC. Gaining evidence on coronary inflammation. J Cardiovasc Comput Tomogr 2021; 15:455-456. [PMID: 33952437 DOI: 10.1016/j.jcct.2021.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 11/23/2022]
Affiliation(s)
- Anna Baritussio
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Azienda Ospedale-Università, University of Padua, Padua, Italy.
| | - Michelle C Williams
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
43
|
Nono Nankam PA, Blüher M. Retinol-binding protein 4 in obesity and metabolic dysfunctions. Mol Cell Endocrinol 2021; 531:111312. [PMID: 33957191 DOI: 10.1016/j.mce.2021.111312] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022]
Abstract
Excessive increased adipose tissue mass in obesity is associated with numerous co-morbid disorders including increased risk of type 2 diabetes, fatty liver disease, hypertension, dyslipidemia, cardiovascular diseases, dementia, airway disease and some cancers. The causal mechanisms explaining these associations are not fully understood. Adipose tissue is an active endocrine organ that secretes many adipokines, cytokines and releases metabolites. These biomolecules referred to as adipocytokines play a significant role in the regulation of whole-body energy homeostasis and metabolism by influencing and altering target tissues function. Understanding the mechanisms of adipocytokine actions represents a hot topic in obesity research. Among several secreted bioactive signalling molecules from adipose tissue and liver, retinol-binding protein 4 (RBP4) has been associated with systemic insulin resistance, dyslipidemia, type 2 diabetes and other metabolic diseases. Here, we aim to review and discuss the current knowledge on RBP4 with a focus on its role in the pathogenesis of obesity comorbid diseases.
Collapse
Affiliation(s)
- Pamela A Nono Nankam
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Germany.
| | - 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, Germany; Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Germany
| |
Collapse
|
44
|
Kalafati M, Lenz M, Ertaylan G, Arts ICW, Evelo CT, van Greevenbroek MMJ, Blaak EE, Adriaens M, Kutmon M. Assessing the Contribution of Relative Macrophage Frequencies to Subcutaneous Adipose Tissue. Front Nutr 2021; 8:675935. [PMID: 34136521 PMCID: PMC8200404 DOI: 10.3389/fnut.2021.675935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/16/2021] [Indexed: 01/09/2023] Open
Abstract
Background: Macrophages play an important role in regulating adipose tissue function, while their frequencies in adipose tissue vary between individuals. Adipose tissue infiltration by high frequencies of macrophages has been linked to changes in adipokine levels and low-grade inflammation, frequently associated with the progression of obesity. The objective of this project was to assess the contribution of relative macrophage frequencies to the overall subcutaneous adipose tissue gene expression using publicly available datasets. Methods: Seven publicly available microarray gene expression datasets from human subcutaneous adipose tissue biopsies (n = 519) were used together with TissueDecoder to determine the adipose tissue cell-type composition of each sample. We divided the subjects in four groups based on their relative macrophage frequencies. Differential gene expression analysis between the high and low relative macrophage frequencies groups was performed, adjusting for sex and study. Finally, biological processes were identified using pathway enrichment and network analysis. Results: We observed lower frequencies of adipocytes and higher frequencies of adipose stem cells in individuals characterized by high macrophage frequencies. We additionally studied whether, within subcutaneous adipose tissue, interindividual differences in the relative frequencies of macrophages were reflected in transcriptional differences in metabolic and inflammatory pathways. Adipose tissue of individuals with high macrophage frequencies had a higher expression of genes involved in complement activation, chemotaxis, focal adhesion, and oxidative stress. Similarly, we observed a lower expression of genes involved in lipid metabolism, fatty acid synthesis, and oxidation and mitochondrial respiration. Conclusion: We present an approach that combines publicly available subcutaneous adipose tissue gene expression datasets with a deconvolution algorithm to calculate subcutaneous adipose tissue cell-type composition. The results showed the expected increased inflammation gene expression profile accompanied by decreased gene expression in pathways related to lipid metabolism and mitochondrial respiration in subcutaneous adipose tissue in individuals characterized by high macrophage frequencies. This approach demonstrates the hidden strength of reusing publicly available data to gain cell-type-specific insights into adipose tissue function.
Collapse
Affiliation(s)
- Marianthi Kalafati
- Deparment of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Michael Lenz
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Institute of Organismic and Molecular Evolution, Johannes Gutenberg University of Mainz, Mainz, Germany.,Preventive Cardiology and Preventive Medicine-Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Gökhan Ertaylan
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Unit Health, Flemish Institute for Technological Research, Antwerp, Belgium
| | - Ilja C W Arts
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Department of Epidemiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Chris T Evelo
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Department of Bioinformatics-BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Ellen E Blaak
- Deparment of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Michiel Adriaens
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| | - Martina Kutmon
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Department of Bioinformatics-BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
45
|
Contribution of Adipose Tissue Oxidative Stress to Obesity-Associated Diabetes Risk and Ethnic Differences: Focus on Women of African Ancestry. Antioxidants (Basel) 2021; 10:antiox10040622. [PMID: 33921645 PMCID: PMC8073769 DOI: 10.3390/antiox10040622] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 12/14/2022] Open
Abstract
Adipose tissue (AT) storage capacity is central in the maintenance of whole-body homeostasis, especially in obesity states. However, sustained nutrients overflow may dysregulate this function resulting in adipocytes hypertrophy, AT hypoxia, inflammation and oxidative stress. Systemic inflammation may also contribute to the disruption of AT redox equilibrium. AT and systemic oxidative stress have been involved in the development of obesity-associated insulin resistance (IR) and type 2 diabetes (T2D) through several mechanisms. Interestingly, fat accumulation, body fat distribution and the degree of how adiposity translates into cardio-metabolic diseases differ between ethnicities. Populations of African ancestry have a higher prevalence of obesity and higher T2D risk than populations of European ancestry, mainly driven by higher rates among African women. Considering the reported ethnic-specific differences in AT distribution and function and higher levels of systemic oxidative stress markers, oxidative stress is a potential contributor to the higher susceptibility for metabolic diseases in African women. This review summarizes existing evidence supporting this hypothesis while acknowledging a lack of data on AT oxidative stress in relation to IR in Africans, and the potential influence of other ethnicity-related modulators (e.g., genetic-environment interplay, socioeconomic factors) for consideration in future studies with different ethnicities.
Collapse
|
46
|
Piccotti F, Rybinska I, Scoccia E, Morasso C, Ricciardi A, Signati L, Triulzi T, Corsi F, Truffi M. Lipofilling in Breast Oncological Surgery: A Safe Opportunity or Risk for Cancer Recurrence? Int J Mol Sci 2021; 22:ijms22073737. [PMID: 33916703 PMCID: PMC8038405 DOI: 10.3390/ijms22073737] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 02/08/2023] Open
Abstract
Lipofilling (LF) is a largely employed technique in reconstructive and esthetic breast surgery. Over the years, it has demonstrated to be extremely useful for treatment of soft tissue defects after demolitive or conservative breast cancer surgery and different procedures have been developed to improve the survival of transplanted fat graft. The regenerative potential of LF is attributed to the multipotent stem cells found in large quantity in adipose tissue. However, a growing body of pre-clinical evidence shows that adipocytes and adipose-derived stromal cells may have pro-tumorigenic potential. Despite no clear indication from clinical studies has demonstrated an increased risk of cancer recurrence upon LF, these observations challenge the oncologic safety of the procedure. This review aims to provide an updated overview of both the clinical and the pre-clinical indications to the suitability and safety of LF in breast oncological surgery. Cellular and molecular players in the crosstalk between adipose tissue and cancer are described, and heterogeneous contradictory results are discussed, highlighting that important issues still remain to be solved to get a clear understanding of LF safety in breast cancer patients.
Collapse
Affiliation(s)
- Francesca Piccotti
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
| | - Ilona Rybinska
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (T.T.)
| | - Elisabetta Scoccia
- Breast Unit, Surgery Department, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (E.S.); (F.C.)
| | - Carlo Morasso
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
| | - Alessandra Ricciardi
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
| | - Lorena Signati
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università Degli Studi di Milano, 20157 Milano, Italy;
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (T.T.)
| | - Fabio Corsi
- Breast Unit, Surgery Department, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (E.S.); (F.C.)
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università Degli Studi di Milano, 20157 Milano, Italy;
| | - Marta Truffi
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
- Correspondence: ; Tel.: +39-0382-592219
| |
Collapse
|
47
|
Sabbatini M, Faruggio S, Verna G, Magnelli V, Dondero F, Boldorini R, Cannas M, Grossini E. Processing Adipose Tissue to Make it More Stable When Used for Refilling: A Morphologic and Immunohistochemistry Evaluation. INQUIRY : A JOURNAL OF MEDICAL CARE ORGANIZATION, PROVISION AND FINANCING 2021; 58:469580211061030. [PMID: 34894844 PMCID: PMC8679401 DOI: 10.1177/00469580211061030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Breast reconstruction has gained from lipofilling the possibility to recover the aesthetic outcome of anatomical profile in a more natural appearance. However, until today, the long-term graft survival remains unpredictable, and sometimes it does not guarantee a well-adequate aesthetic result. In the present work, the morphological changes, occurring in fat mass used for refilling, harvested by the Coleman's procedure or through the washing/fragmenting procedure were analysed. Adipocyte size; immunohistochemistry against CD8, CD31, CD68 and M2-type macrophages and catalase enzyme, were analysed in vitro on fat mass cultured for 4 weeks. Our observation reveals an increase of connective tissue around the mass and a high number of immune cells occurrence in fat mass harvested by the Coleman's procedure. Instead, the washing/fragmented procedure would reduce the number of immune cells within the fat mass, increase the size of adipocytes, and cause a wider presence of active vessels profile and greater catalase expression. We hypothesize that the fat mass processed by the Coleman's procedure would remain more reactive due to a higher number of immune and macrophages cells, prone to develop cystic formation, leading to a suboptimal integration in the recipient site. On the other hand, the conditions more prone to realize an optimal integration would occur in the fat mass processed by the washing/fragmenting procedure: a reduced number of immune cells, low amount of connective tissue, presence of larger adipocytes. Follow-up monitoring did support our conclusion, as we observed a reduction of re-intervention for refilling procedure in patients treated with the washing/fragmenting procedure.
Collapse
Affiliation(s)
- Maurizio Sabbatini
- Department of Science and
Technology Innovation, UPO University, Alessandria, Italy
| | - Serena Faruggio
- Department of Translational
Medicine, UPO University, Novara, Italy
| | - Giovanni Verna
- Department of Plastic and
Reconstructive Surgery, Hospital “Maggiore Della
Carità,” Novara, Italy
| | - Valeria Magnelli
- Department of Science and
Technology Innovation, UPO University, Alessandria, Italy
| | - Francesco Dondero
- Department of Science and
Technology Innovation, UPO University, Alessandria, Italy
| | - Renzo Boldorini
- Department of Health Science,
Section of Pathological Anatomy, UPO University, Novara, Italy
| | - Mario Cannas
- Department of Health Sciences, UPO University, Novara, Italy
| | - Elena Grossini
- Department of Translational
Medicine, UPO University, Novara, Italy
| |
Collapse
|
48
|
Orsso CE, Colin-Ramirez E, Field CJ, Madsen KL, Prado CM, Haqq AM. Adipose Tissue Development and Expansion from the Womb to Adolescence: An Overview. Nutrients 2020; 12:E2735. [PMID: 32911676 PMCID: PMC7551046 DOI: 10.3390/nu12092735] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/29/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
Prevalence rates of pediatric obesity continue to rise worldwide. Adipose tissue (AT) development and expansion initiate in the fetus and extend throughout the lifespan. This paper presents an overview of the AT developmental trajectories from the intrauterine period to adolescence; factors determining adiposity expansion are also discussed. The greatest fetal increases in AT were observed in the third pregnancy trimester, with growing evidence suggesting that maternal health and nutrition, toxin exposure, and genetic defects impact AT development. From birth up to six months, healthy term newborns experience steep increases in AT; but a subsequent reduction in AT is observed during infancy. Important determinants of AT in infancy identified in this review included feeding practices and factors shaping the gut microbiome. Low AT accrual rates are maintained up to puberty onset, at which time, the pattern of adiposity expansion becomes sex dependent. As girls experience rapid increases and boys experience decreases in AT, sexual dimorphism in hormone secretion can be considered the main contributor for changes. Eating patterns/behaviors and interactions between dietary components, gut microbiome, and immune cells also influence AT expansion. Despite the plasticity of this tissue, substantial evidence supports that adiposity at birth and infancy highly influences its levels across subsequent life stages. Thus, a unique window of opportunity for the prevention and/or slowing down of the predisposition toward obesity, exists from pregnancy through childhood.
Collapse
Affiliation(s)
- Camila E. Orsso
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; (C.E.O.); (C.J.F.); (C.M.P.)
| | | | - Catherine J. Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; (C.E.O.); (C.J.F.); (C.M.P.)
| | - Karen L. Madsen
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2C2, Canada;
| | - Carla M. Prado
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; (C.E.O.); (C.J.F.); (C.M.P.)
| | - Andrea M. Haqq
- Department of Pediatrics and Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R7, Canada
| |
Collapse
|
49
|
Defour M, Michielsen CCJR, O'Donovan SD, Afman LA, Kersten S. Transcriptomic signature of fasting in human adipose tissue. Physiol Genomics 2020; 52:451-467. [PMID: 32866087 DOI: 10.1152/physiolgenomics.00083.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Little is known about gene regulation by fasting in human adipose tissue. Accordingly, the objective of this study was to investigate the effects of fasting on adipose tissue gene expression in humans. To that end, subcutaneous adipose tissue biopsies were collected from 11 volunteers 2 and 26 h after consumption of a standardized meal. For comparison, epididymal adipose tissue was collected from C57Bl/6J mice in the ab libitum-fed state and after a 16 h fast. The timing of sampling adipose tissue roughly corresponds with the near depletion of liver glycogen. Transcriptome analysis was carried out using Affymetrix microarrays. We found that, 1) fasting downregulated numerous metabolic pathways in human adipose tissue, including triglyceride and fatty acid synthesis, glycolysis and glycogen synthesis, TCA cycle, oxidative phosphorylation, mitochondrial translation, and insulin signaling; 2) fasting downregulated genes involved in proteasomal degradation in human adipose tissue; 3) fasting had much less pronounced effects on the adipose tissue transcriptome in humans than mice; 4) although major overlap in fasting-induced gene regulation was observed between human and mouse adipose tissue, many genes were differentially regulated in the two species, including genes involved in insulin signaling (PRKAG2, PFKFB3), PPAR signaling (PPARG, ACSL1, HMGCS2, SLC22A5, ACOT1), glycogen metabolism (PCK1, PYGB), and lipid droplets (PLIN1, PNPLA2, CIDEA, CIDEC). In conclusion, although numerous genes and pathways are regulated similarly by fasting in human and mouse adipose tissue, many genes show very distinct responses to fasting in humans and mice. Our data provide a useful resource to study adipose tissue function during fasting.
Collapse
Affiliation(s)
- Merel Defour
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Charlotte C J R Michielsen
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Shauna D O'Donovan
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Lydia A Afman
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|