1
|
Lecoutre S, Rebière C, Maqdasy S, Lambert M, Dussaud S, Abatan JB, Dugail I, Gautier EL, Clément K, Marcelin G. Enhancing adipose tissue plasticity: progenitor cell roles in metabolic health. Nat Rev Endocrinol 2025:10.1038/s41574-024-01071-y. [PMID: 39757324 DOI: 10.1038/s41574-024-01071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 01/07/2025]
Abstract
Adipose tissue demonstrates considerable plasticity and heterogeneity, enabling metabolic, cellular and structural adaptations to environmental signals. This adaptability is key for maintaining metabolic homeostasis. Impaired adipose tissue plasticity can lead to abnormal adipose tissue responses to metabolic cues, which contributes to the development of cardiometabolic diseases. In chronic obesity, white adipose tissue undergoes pathological remodelling marked by adipocyte hypertrophy, chronic inflammation and fibrosis, which are linked to local and systemic insulin resistance. Research data suggest that the capacity for healthy or unhealthy white adipose tissue remodelling might depend on the intrinsic diversity of adipose progenitor cells (APCs), which sense and respond to metabolic cues. This Review highlights studies on APCs as key determinants of adipose tissue plasticity, discussing differences between subcutaneous and visceral adipose tissue depots during development, growth and obesity. Modulating APC functions could improve strategies for treating adipose tissue dysfunction and metabolic diseases in obesity.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| | - Clémentine Rebière
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Salwan Maqdasy
- Department of Medicine, Karolinska Institutet Hospital, Stockholm, Sweden
| | - Mélanie Lambert
- Institut National de la Santé et de la Recherche Médicale, Bobigny, France
- Labex Inflamex, Université Sorbonne Paris Nord, Alliance Sorbonne Paris Cité, Bobigny, France
| | - Sébastien Dussaud
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Jimon Boniface Abatan
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Emmanuel L Gautier
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
- Department of Nutrition, Pitie-Salpêtriere Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Geneviève Marcelin
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| |
Collapse
|
2
|
Solsona-Vilarrasa E, Vousden KH. Obesity, white adipose tissue and cancer. FEBS J 2024. [PMID: 39496581 DOI: 10.1111/febs.17312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024]
Abstract
White adipose tissue (WAT) is crucial for whole-body energy homeostasis and plays an important role in metabolic and hormonal regulation. While healthy WAT undergoes controlled expansion and contraction to meet the body's requirements, dysfunctional WAT in conditions like obesity is characterized by excessive tissue expansion, alterations in lipid homeostasis, inflammation, hypoxia, and fibrosis. Obesity is strongly associated with an increased risk of numerous cancers, with obesity-induced WAT dysfunction influencing cancer development through various mechanisms involving both systemic and local interactions between adipose tissue and tumors. Unhealthy obese WAT affects circulating levels of free fatty acids and factors like leptin, adiponectin, and insulin, altering systemic lipid metabolism and inducing inflammation that supports tumor growth. Similar mechanisms are observed locally in an adipose-rich tumor microenvironment (TME), where WAT cells can also trigger extracellular matrix remodeling, thereby enhancing the TME's ability to promote tumor growth. Moreover, tumors reciprocally interact with WAT, creating a bidirectional communication that further enhances tumorigenesis. This review focuses on the complex interplay between obesity, WAT dysfunction, and primary tumor growth, highlighting potential targets for therapeutic intervention.
Collapse
|
3
|
Ellis CPS, Tero BW, Potts CM, Malka KT, Yang X, Hamilton J, Vary C, Khalil A, Liaw L. Cellular Characteristics and Protein Signatures of Human Adipose Tissues from Donors With or Without Advanced Coronary Artery Disease. Biomedicines 2024; 12:2453. [PMID: 39595019 PMCID: PMC11592159 DOI: 10.3390/biomedicines12112453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Perivascular adipose tissue (PVAT) exerts a paracrine effect on blood vessels and our objective was to understand PVAT molecular signatures related to cardiovascular disease. Methods: We studied two groups: those undergoing mitral valve repair/replacement (VR, n = 16) and coronary artery bypass graft (CABG, n = 38). VR donors did not have coronary artery disease, whereas CABG donors had advanced coronary artery disease. Clinical and tissue pathologies and proteomics from adipose tissue were assessed. Results: Donors undergoing VR had a lower body mass index (p = 0.01), HbA1C (p = 0.0023), and incidence of diabetes (p = 0.022) compared to CABG. VR donors were overall healthier, with higher cardiac function compared to CABG donors, based on ejection fraction. Although adipose histopathology between groups was not markedly different, PVAT had smaller and more adipocytes compared to subcutaneous adipose tissues. These differences were validated by whole specimen automated morphological analysis, and anisotropy analysis showed small (2.8-7.5 μm) and large (22.8-64.4 μm) scale differences between perivascular and subcutaneous adipose tissue from CABG donors, and small scale changes (2.8-7.5 μm) between perivascular and subcutaneous adipose tissue from VR donors. Distinct protein signatures in PVAT and subcutaneous adipose tissue include those involved in secretion, exosomes and vesicles, insulin resistance, and adipocyte identity. Comparing PVAT and subcutaneous adipose tissue from CABG donors, there were 82 significantly different proteins identified with log fold change ≥ 0.3 or ≤-0.3 (p < 0.05). Using this threshold, there were 36 differences when comparing PVAT and subcutaneous adipose tissue from VR donors, 58 differences when comparing PVAT from CABG or VR donors, and 55 when comparing subcutaneous adipose tissue from CABG vs. VR donors. Conclusions: Routine histopathology cannot differentiate between PVAT from donors with or without coronary artery disease, but multiscale anisotropy analysis discriminated between these populations. Our mass spectrometry analysis identified a cohort of proteins that distinguish between adipose depots, and are also associated with the presence or absence of coronary artery disease.
Collapse
Affiliation(s)
- Caitlin P. S. Ellis
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
| | - Benjamin W. Tero
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Christian M. Potts
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Kimberly T. Malka
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Xuehui Yang
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Joshua Hamilton
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
- CompuMAINE Lab, Department of Chemical and Biomedical Engineering, University of Maine, Orono, ME 04469, USA
| | - Calvin Vary
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
| | - Andre Khalil
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
- CompuMAINE Lab, Department of Chemical and Biomedical Engineering, University of Maine, Orono, ME 04469, USA
| | - Lucy Liaw
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
| |
Collapse
|
4
|
Greig JC, Tipping WJ, Graham D, Faulds K, Gould GW. New insights into lipid and fatty acid metabolism from Raman spectroscopy. Analyst 2024. [PMID: 39258960 DOI: 10.1039/d4an00846d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
One of the challenges facing biology is to understand metabolic events at a single cellular level. While approaches to examine dynamics of protein distribution or report on spatiotemporal location of signalling molecules are well-established, tools for the dissection of metabolism in single living cells are less common. Advances in Raman spectroscopy, such as stimulated Raman scattering (SRS), are beginning to offer new insights into metabolic events in a range of experimental systems, including model organisms and clinical samples, and across a range of disciplines. Despite the power of Raman imaging, it remains a relatively under-used technique to approach biological problems, in part because of the specialised nature of the analysis. To raise the profile of this method, here we consider some key studies which illustrate how Raman spectroscopy has revealed new insights into fatty acid and lipid metabolism across a range of cellular systems. The powerful and non-invasive nature of this approach offers a new suite of tools for biomolecular scientists to address how metabolic events within cells informs on or underpins biological function. We illustrate potential biological applications, discuss some recent advances, and offer a direction of travel for metabolic research in this area.
Collapse
Affiliation(s)
- Justin C Greig
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, UK.
| | | | - Duncan Graham
- Pure and Applied Chemistry, University of Strathclyde, UK
| | - Karen Faulds
- Pure and Applied Chemistry, University of Strathclyde, UK
| | - Gwyn W Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, UK.
| |
Collapse
|
5
|
González-Garibay AS, Sandoval G, Torres-González OR, Bastidas-Ramírez BE, Sánchez-Hernández IM, Padilla-Camberos E. Agave-Laurate-Bioconjugated Fructans Decrease Hyperinsulinemia and Insulin Resistance, Whilst Increasing IL-10 in Rats with Metabolic Syndrome Induced by a High-Fat Diet. Pharmaceuticals (Basel) 2024; 17:1036. [PMID: 39204141 PMCID: PMC11357657 DOI: 10.3390/ph17081036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Metabolic syndrome (MetS) comprises a cluster of metabolic risk factors, which include obesity, hypertriglyceridemia, high blood pressure, and insulin resistance. The purpose of this study was to evaluate the effects of laurate-bioconjugated fructans on pro- and anti-inflammatory cytokines in Wistar rats with MetS induced by a high-fat diet. Laurate-bioconjugated fructans were synthesized with agave fructans, immobilized lipase B, and vinyl laureate as the acylant. Groups were fed a standard diet (NORMAL), a high-fat diet (HFD), or a high-fat diet plus laurate-bioconjugated fructans (FL PREV) for 9 weeks. A fourth group received a high-fat diet for 6 weeks, followed by simultaneous exposure to a high-fat diet and laurate-bioconjugated fructans for 3 additional weeks (FL REV). The dose of laurate-bioconjugated fructans was 130 mg/kg. Laurate-bioconjugated fructans reduced food and energy intake, body weight, body mass index, abdominal circumference, adipose tissue, adipocyte area, serum triglycerides, insulin, insulin resistance, and C-reactive protein but they increased IL-10 protein serum levels and mRNA expression. The impact of laurate-bioconjugated fructans on zoometric and metabolic parameters supports their potential as therapeutic agents to improve obesity, obesity comorbidities, insulin resistance, type 2 diabetes mellitus, and MetS.
Collapse
Affiliation(s)
- Angélica Sofía González-Garibay
- Medical and Pharmaceutical Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Av. Normalistas No. 800 Col. Colinas de la Normal, Guadalajara C.P. 44270, Jalisco, Mexico
- Department of Molecular Biology and Genomics, Institute of Research on Chronic Degenerative Diseases, University Center of Health Sciences, Universidad de Guadalajara, Sierra Mojada No. 950 Col. Independencia, Guadalajara C.P. 44340, Jalisco, Mexico
| | - Georgina Sandoval
- Medical and Pharmaceutical Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Av. Normalistas No. 800 Col. Colinas de la Normal, Guadalajara C.P. 44270, Jalisco, Mexico
| | - Omar Ricardo Torres-González
- Medical and Pharmaceutical Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Av. Normalistas No. 800 Col. Colinas de la Normal, Guadalajara C.P. 44270, Jalisco, Mexico
| | - Blanca Estela Bastidas-Ramírez
- Department of Molecular Biology and Genomics, Institute of Research on Chronic Degenerative Diseases, University Center of Health Sciences, Universidad de Guadalajara, Sierra Mojada No. 950 Col. Independencia, Guadalajara C.P. 44340, Jalisco, Mexico
| | - Iván Moisés Sánchez-Hernández
- Medical and Pharmaceutical Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Av. Normalistas No. 800 Col. Colinas de la Normal, Guadalajara C.P. 44270, Jalisco, Mexico
| | - Eduardo Padilla-Camberos
- Medical and Pharmaceutical Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Av. Normalistas No. 800 Col. Colinas de la Normal, Guadalajara C.P. 44270, Jalisco, Mexico
| |
Collapse
|
6
|
Pagani A, Duscher D, Kempa S, Ghods M, Prantl L. Preliminary Single-Cell RNA-Sequencing Analysis Uncovers Adipocyte Heterogeneity in Lipedema. Cells 2024; 13:1028. [PMID: 38920656 PMCID: PMC11201579 DOI: 10.3390/cells13121028] [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: 04/28/2024] [Revised: 06/08/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024] Open
Abstract
Background: Despite its increasing incidence and prevalence throughout Western countries, lipedema continues to be a very enigmatic disease, often misunderstood or misdiagnosed by the medical community and with an intrinsic pathology that is difficult to trace. The nature of lipedemic tissue is one of hypertrophic adipocytes and poor tissue turnover. So far, there are no identified pathways responsible, and little is known about the cell populations of lipedemic fat. Methods: Adipose tissue samples were collected from affected areas of both lipedema and healthy participants. For single-cell RNA sequencing analysis, the samples were dissociated into single-cell suspensions using enzymatic digestion and then encapsulated into nanoliter-sized droplets containing barcoded beads. Within each droplet, cellular mRNA was converted into complementary DNA. Complementary DNA molecules were then amplified for downstream analysis. Results: The single-cell RNA-sequencing analysis revealed three distinct adipocyte populations at play in lipedema. These populations have unique gene signatures which can be characterized as a lipid generating adipocyte, a disease catalyst adipocyte, and a lipedemic adipocyte. Conclusions: The single-cell RNA sequencing of lipedemic tissue samples highlights a triad of distinct adipocyte subpopulations, each characterized by unique gene signatures and functional roles. The interplay between these adipocyte subtypes offers promising insights into the complex pathophysiology of lipedema.
Collapse
Affiliation(s)
- Andrea Pagani
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany
| | - Dominik Duscher
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany
| | - Sally Kempa
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany
| | - Mojtaba Ghods
- Department of Plastic, Aesthetic and Reconstructive Surgery, Clinic Ernst von Bergmann, Charlottenstraße 71, 14467 Potsdam, Germany
| | - Lukas Prantl
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
7
|
Okudzhava L, Schulz S, Fischi‐Gomez E, Girard G, Machann J, Koch PJ, Thiran J, Münte TF, Heldmann M. White adipose tissue distribution and amount are associated with increased white matter connectivity. Hum Brain Mapp 2024; 45:e26654. [PMID: 38520361 PMCID: PMC10960552 DOI: 10.1002/hbm.26654] [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: 10/31/2023] [Revised: 02/09/2024] [Accepted: 02/27/2024] [Indexed: 03/25/2024] Open
Abstract
Obesity represents a significant public health concern and is linked to various comorbidities and cognitive impairments. Previous research indicates that elevated body mass index (BMI) is associated with structural changes in white matter (WM). However, a deeper characterization of body composition is required, especially considering the links between abdominal obesity and metabolic dysfunction. This study aims to enhance our understanding of the relationship between obesity and WM connectivity by directly assessing the amount and distribution of fat tissue. Whole-body magnetic resonance imaging (MRI) was employed to evaluate total adipose tissue (TAT), visceral adipose tissue (VAT), and subcutaneous adipose tissue (SAT), while MR liver spectroscopy measured liver fat content in 63 normal-weight, overweight, and obese males. WM connectivity was quantified using microstructure-informed tractography. Connectome-based predictive modeling was used to predict body composition metrics based on WM connectomes. Our analysis revealed a positive dependency between BMI, TAT, SAT, and WM connectivity in brain regions involved in reward processing and appetite regulation, such as the insula, nucleus accumbens, and orbitofrontal cortex. Increased connectivity was also observed in cognitive control and inhibition networks, including the middle frontal gyrus and anterior cingulate cortex. No significant associations were found between WM connectivity and VAT or liver fat. Our findings suggest that altered neural communication between these brain regions may affect cognitive processes, emotional regulation, and reward perception in individuals with obesity, potentially contributing to weight gain. While our study did not identify a link between WM connectivity and VAT or liver fat, further investigation of the role of various fat depots and metabolic factors in brain networks is required to advance obesity prevention and treatment approaches.
Collapse
Affiliation(s)
- Liana Okudzhava
- Department of NeurologyUniversity of LübeckLübeckGermany
- Center of Brain, Behavior and MetabolismUniversity of LübeckLübeckGermany
| | - Stephanie Schulz
- Department of NeurologyUniversity of LübeckLübeckGermany
- Center of Brain, Behavior and MetabolismUniversity of LübeckLübeckGermany
| | - Elda Fischi‐Gomez
- CIBM Center for Biomedical ImagingLausanneSwitzerland
- Radiology DepartmentLausanne University and University Hospital (CHUV)LausanneSwitzerland
- Signal Processing Laboratory (LTS5), Ecole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Gabriel Girard
- CIBM Center for Biomedical ImagingLausanneSwitzerland
- Department of Computer ScienceUniversité de SherbrookeSherbrookeQuebecCanada
| | - Jürgen Machann
- Section on Experimental Radiology, Department of RadiologyEberhard‐Karls UniversityTübingenGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center MunichUniversity of TübingenTübingenGermany
| | - Philipp J. Koch
- Department of NeurologyUniversity of LübeckLübeckGermany
- Center of Brain, Behavior and MetabolismUniversity of LübeckLübeckGermany
| | - Jean‐Philippe Thiran
- CIBM Center for Biomedical ImagingLausanneSwitzerland
- Radiology DepartmentLausanne University and University Hospital (CHUV)LausanneSwitzerland
- Signal Processing Laboratory (LTS5), Ecole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Thomas F. Münte
- Department of NeurologyUniversity of LübeckLübeckGermany
- Center of Brain, Behavior and MetabolismUniversity of LübeckLübeckGermany
| | - Marcus Heldmann
- Department of NeurologyUniversity of LübeckLübeckGermany
- Center of Brain, Behavior and MetabolismUniversity of LübeckLübeckGermany
- Institute of Psychology IIUniversity of LübeckLübeckGermany
| |
Collapse
|
8
|
Cani PD, Van Hul M. Gut microbiota in overweight and obesity: crosstalk with adipose tissue. Nat Rev Gastroenterol Hepatol 2024; 21:164-183. [PMID: 38066102 DOI: 10.1038/s41575-023-00867-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 03/02/2024]
Abstract
Overweight and obesity are characterized by excessive fat mass accumulation produced when energy intake exceeds energy expenditure. One plausible way to control energy expenditure is to modulate thermogenic pathways in white adipose tissue (WAT) and/or brown adipose tissue (BAT). Among the different environmental factors capable of influencing host metabolism and energy balance, the gut microbiota is now considered a key player. Following pioneering studies showing that mice lacking gut microbes (that is, germ-free mice) or depleted of their gut microbiota (that is, using antibiotics) developed less adipose tissue, numerous studies have investigated the complex interactions existing between gut bacteria, some of their membrane components (that is, lipopolysaccharides), and their metabolites (that is, short-chain fatty acids, endocannabinoids, bile acids, aryl hydrocarbon receptor ligands and tryptophan derivatives) as well as their contribution to the browning and/or beiging of WAT and changes in BAT activity. In this Review, we discuss the general physiology of both WAT and BAT. Subsequently, we introduce how gut bacteria and different microbiota-derived metabolites, their receptors and signalling pathways can regulate the development of adipose tissue and its metabolic capacities. Finally, we describe the key challenges in moving from bench to bedside by presenting specific key examples.
Collapse
Affiliation(s)
- Patrice D Cani
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
9
|
Bettinetti-Luque M, Trujillo-Estrada L, Garcia-Fuentes E, Andreo-Lopez J, Sanchez-Varo R, Garrido-Sánchez L, Gómez-Mediavilla Á, López MG, Garcia-Caballero M, Gutierrez A, Baglietto-Vargas D. Adipose tissue as a therapeutic target for vascular damage in Alzheimer's disease. Br J Pharmacol 2024; 181:840-878. [PMID: 37706346 DOI: 10.1111/bph.16243] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/15/2023] Open
Abstract
Adipose tissue has recently been recognized as an important endocrine organ that plays a crucial role in energy metabolism and in the immune response in many metabolic tissues. With this regard, emerging evidence indicates that an important crosstalk exists between the adipose tissue and the brain. However, the contribution of adipose tissue to the development of age-related diseases, including Alzheimer's disease, remains poorly defined. New studies suggest that the adipose tissue modulates brain function through a range of endogenous biologically active factors known as adipokines, which can cross the blood-brain barrier to reach the target areas in the brain or to regulate the function of the blood-brain barrier. In this review, we discuss the effects of several adipokines on the physiology of the blood-brain barrier, their contribution to the development of Alzheimer's disease and their therapeutic potential. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
- Miriam Bettinetti-Luque
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Laura Trujillo-Estrada
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Eduardo Garcia-Fuentes
- Unidad de Gestión Clínica Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
- CIBER de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Juana Andreo-Lopez
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Raquel Sanchez-Varo
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Lourdes Garrido-Sánchez
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
| | - Ángela Gómez-Mediavilla
- Departamento de Farmacología, Facultad de Medicina. Instituto Teófilo Hernando para la I+D de Fármacos, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuela G López
- Departamento de Farmacología, Facultad de Medicina. Instituto Teófilo Hernando para la I+D de Fármacos, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Sanitarias (IIS-IP), Hospital Universitario de la Princesa, Madrid, Spain
| | - Melissa Garcia-Caballero
- Departamento de Biología Molecular y Bioquímica, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Antonia Gutierrez
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - David Baglietto-Vargas
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
10
|
Abujrais S, Ubhayasekera SJKA, Bergquist J. Analysis of tryptophan metabolites and related compounds in human and murine tissue: development and validation of a quantitative and semi-quantitative method using high resolution mass spectrometry. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:1074-1082. [PMID: 38282545 DOI: 10.1039/d3ay01959d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
This study explores the metabolic differences between human and murine plasma in addition to differences between murine subcutaneous and visceral white adipose tissue. A quantitative and semi-quantitative targeted method was developed and validated for this purpose. The quantitative method includes tryptophan and its metabolites in addition to tyrosine, phenylalanine, taurine, B vitamins, neopterin, cystathionine and hypoxanthine. While the semi-quantitative method includes; 3-indoleacetic acid, 5-hydroxyindoleacetic acid, acetylcholine, asymmetric dimethylarginine, citrulline and methionine. Sample preparation was based on protein precipitation, while quantification was conducted using ultrahigh-performance liquid chromatography coupled to a quadrupole Orbitrap tandem mass spectrometer with electrospray ionization in the parallel reaction monitoring (PRM) mode. The low limit of quantification for all metabolites ranged from 1 to 200 ng mL-1. Matrix effects and recoveries for stable isotope labelled internal standards were evaluated, with most having a coefficient of variation (CV) of less than 15%. Results showed that a majority of the analytes passed both the intra- and interday precision and accuracy criteria. The comparative analysis of human and murine plasma metabolites reveals species-specific variations within the tryptophan metabolic pathway. Notably, murine plasma generally exhibits elevated concentrations of most compounds in this pathway, with the exceptions of kynurenine and quinolinic acid. Moreover, the investigation uncovers noteworthy metabolic disparities between murine visceral and subcutaneous white adipose tissues, with the subcutaneous tissue demonstrating significantly higher concentrations of tryptophan, phenylalanine, tyrosine, and serotonin. The findings also show that even a semi-quantitative method can provide comparable results to quantitative methods from other studies and be effective for assessing metabolites in a complex sample. Overall, this study provides a robust platform to compare human and murine metabolism, providing a valuable insight to future investigations.
Collapse
Affiliation(s)
- Sandy Abujrais
- Analytical Chemistry and Neurochemistry, Department of Chemistry - BMC, Uppsala University, Box 599, 75124, Uppsala, Sweden.
- The ME/CFS Collaborative Research Centre at Uppsala University, Sweden
| | - S J Kumari A Ubhayasekera
- Analytical Chemistry and Neurochemistry, Department of Chemistry - BMC, Uppsala University, Box 599, 75124, Uppsala, Sweden.
- The ME/CFS Collaborative Research Centre at Uppsala University, Sweden
| | - Jonas Bergquist
- Analytical Chemistry and Neurochemistry, Department of Chemistry - BMC, Uppsala University, Box 599, 75124, Uppsala, Sweden.
- The ME/CFS Collaborative Research Centre at Uppsala University, Sweden
| |
Collapse
|
11
|
Redruello-Romero A, Benitez-Cantos MS, Lopez-Perez D, García-Rubio J, Tamayo F, Pérez-Bartivas D, Moreno-SanJuan S, Ruiz-Palmero I, Puentes-Pardo JD, Vilchez JR, López-Nevot MÁ, García F, Cano C, León J, Carazo Á. Human adipose tissue as a major reservoir of cytomegalovirus-reactive T cells. Front Immunol 2023; 14:1303724. [PMID: 38053998 PMCID: PMC10694288 DOI: 10.3389/fimmu.2023.1303724] [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: 09/28/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
Introduction Cytomegalovirus (CMV) is a common herpesvirus with a high prevalence worldwide. After the acute infection phase, CMV can remain latent in several tissues. CD8 T cells in the lungs and salivary glands mainly control its reactivation control. White adipose tissue (WAT) contains a significant population of memory T cells reactive to viral antigens, but CMV specificity has mainly been studied in mouse WAT. Therefore, we obtained blood, omental WAT (oWAT), subcutaneous WAT (sWAT), and liver samples from 11 obese donors to characterize the human WAT adaptive immune landscape from a phenotypic and immune receptor specificity perspective. Methods We performed high-throughput sequencing of the T cell receptor (TCR) locus to analyze tissue and blood TCR repertoires of the 11 donors. The presence of TCRs specific to CMV epitopes was tested through ELISpot assays. Moreover, phenotypic characterization of T cells was carried out through flow cytometry. Results High-throughput sequencing analyses revealed that tissue TCR repertoires in oWAT, sWAT, and liver samples were less diverse and dominated by hyperexpanded clones when compared to blood samples. Additionally, we predicted the presence of TCRs specific to viral epitopes, particularly from CMV, which was confirmed by ELISpot assays. Remarkably, we found that oWAT has a higher proportion of CMV-reactive T cells than blood or sWAT. Finally, flow cytometry analyses indicated that most WAT-infiltrated lymphocytes were tissue-resident effector memory CD8 T cells. Discussion Overall, these findings postulate human oWAT as a major reservoir of CMV-specific T cells, presumably for latent viral reactivation control. This study enhances our understanding of the adaptive immune response in human WAT and highlights its potential role in antiviral defense.
Collapse
Affiliation(s)
| | - Maria S. Benitez-Cantos
- Research Unit, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Granada, Spain
- GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - David Lopez-Perez
- Research Unit, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain
| | | | | | - Daniel Pérez-Bartivas
- Research Unit, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
| | - Sara Moreno-SanJuan
- Research Unit, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
- Cytometry and Microscopy Research Service, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
| | - Isabel Ruiz-Palmero
- Research Unit, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
| | - Jose D. Puentes-Pardo
- Research Unit, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Jose R. Vilchez
- Research Unit, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
- Clinical Analyses and Immunology Unit, Virgen de las Nieves University Hospital, Granada, Spain
| | - Miguel Á. López-Nevot
- Research Unit, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Granada, Spain
- Clinical Analyses and Immunology Unit, Virgen de las Nieves University Hospital, Granada, Spain
| | - Federico García
- Research Unit, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
- Clinical Microbiology Unit, San Cecilio University Hospital, Granada, Spain
- Centro de Investigación Biomédica en Red (CIBER) of Infectious Diseases, Health Institute Carlos III, Madrid, Spain
| | - Carlos Cano
- Department of Computer Science and Artificial Intelligence, University of Granada, Granada, Spain
| | - Josefa León
- Research Unit, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
- Digestive Unit, San Cecilio University Hospital, Granada, Spain
| | - Ángel Carazo
- Research Unit, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
- Clinical Microbiology Unit, San Cecilio University Hospital, Granada, Spain
| |
Collapse
|
12
|
Carrillo AE, Vliora M. Adipose Tissue Metabolism in Response to Food Intake. Nutrients 2023; 15:4811. [PMID: 38004204 PMCID: PMC10675001 DOI: 10.3390/nu15224811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
The quality and quantity of the food we consume have a major impact on our general health and longevity [...].
Collapse
Affiliation(s)
- Andres E. Carrillo
- Department of Exercise Science, School of Health Sciences, Chatham University, Pittsburgh, PA 15232, USA
| | - Maria Vliora
- FAME Laboratory, Department of Physical Education and Sport Science, University of Thessaly, 42100 Trikala, Greece;
| |
Collapse
|
13
|
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
|
14
|
Zhu T, Chen X, Jiang S. Progress and obstacles in transplantation of brown adipose tissue or engineered cells with thermogenic potential for metabolic benefits. Front Endocrinol (Lausanne) 2023; 14:1191278. [PMID: 37265692 PMCID: PMC10230949 DOI: 10.3389/fendo.2023.1191278] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/27/2023] [Indexed: 06/03/2023] Open
Abstract
Transplantation of brown adipose tissue (BAT), engineered thermogenic progenitor cells, and adipocytes have received much attention for the improvement of obesity and metabolic disorders. However, even though the thermogenic and metabolic potential exists early after transplantation, the whitening of the brown fat graft occurs with metabolic function significantly impaired. In this review, specific experiment designs, graft outcomes, and metabolic benefits for the transplantation of BAT or engineered cells will be discussed. The current advancements will offer guidance to further investigation, and the obstacles appearing in previous studies will require innovation of BAT transplantation methods.
Collapse
|
15
|
Ahn B. The Function of MondoA and ChREBP Nutrient-Sensing Factors in Metabolic Disease. Int J Mol Sci 2023; 24:ijms24108811. [PMID: 37240157 DOI: 10.3390/ijms24108811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/12/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity is a major global public health concern associated with an increased risk of many health problems, including type 2 diabetes, heart disease, stroke, and some types of cancer. Obesity is also a critical factor in the development of insulin resistance and type 2 diabetes. Insulin resistance is associated with metabolic inflexibility, which interferes with the body's ability to switch from free fatty acids to carbohydrate substrates, as well as with the ectopic accumulation of triglycerides in non-adipose tissue, such as that of skeletal muscle, the liver, heart, and pancreas. Recent studies have demonstrated that MondoA (MLX-interacting protein or MLXIP) and the carbohydrate response element-binding protein (ChREBP, also known as MLXIPL and MondoB) play crucial roles in the regulation of nutrient metabolism and energy homeostasis in the body. This review summarizes recent advances in elucidating the function of MondoA and ChREBP in insulin resistance and related pathological conditions. This review provides an overview of the mechanisms by which MondoA and ChREBP transcription factors regulate glucose and lipid metabolism in metabolically active organs. Understanding the underlying mechanism of MondoA and ChREBP in insulin resistance and obesity can foster the development of new therapeutic strategies for treating metabolic diseases.
Collapse
Affiliation(s)
- Byungyong Ahn
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 44610, Republic of Korea
| |
Collapse
|
16
|
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
|
17
|
Barthelemy J, Bogard G, Wolowczuk I. Beyond energy balance regulation: The underestimated role of adipose tissues in host defense against pathogens. Front Immunol 2023; 14:1083191. [PMID: 36936928 PMCID: PMC10019896 DOI: 10.3389/fimmu.2023.1083191] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/09/2023] [Indexed: 03/06/2023] Open
Abstract
Although the adipose tissue (AT) is a central metabolic organ in the regulation of whole-body energy homeostasis, it is also an important endocrine and immunological organ. As an endocrine organ, AT secretes a variety of bioactive peptides known as adipokines - some of which have inflammatory and immunoregulatory properties. As an immunological organ, AT contains a broad spectrum of innate and adaptive immune cells that have mostly been studied in the context of obesity. However, overwhelming evidence supports the notion that AT is a genuine immunological effector site, which contains all cell subsets required to induce and generate specific and effective immune responses against pathogens. Indeed, AT was reported to be an immune reservoir in the host's response to infection, and a site of parasitic, bacterial and viral infections. In addition, besides AT's immune cells, preadipocytes and adipocytes were shown to express innate immune receptors, and adipocytes were reported as antigen-presenting cells to regulate T-cell-mediated adaptive immunity. Here we review the current knowledge on the role of AT and AT's immune system in host defense against pathogens. First, we will summarize the main characteristics of AT: type, distribution, function, and extraordinary plasticity. Second, we will describe the intimate contact AT has with lymph nodes and vessels, and AT immune cell composition. Finally, we will present a comprehensive and up-to-date overview of the current research on the contribution of AT to host defense against pathogens, including the respiratory viruses influenza and SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Isabelle Wolowczuk
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019 - UMR 9017 - Center for Infection and Immunity of Lille (CIIL), Lille, France
| |
Collapse
|
18
|
Song J, Farris D, Ariza P, Moorjani S, Varghese M, Blin M, Chen J, Tyrrell D, Zhang M, Singer K, Salmon M, Goldstein DR. Age-associated adipose tissue inflammation promotes monocyte chemotaxis and enhances atherosclerosis. Aging Cell 2023; 22:e13783. [PMID: 36683460 PMCID: PMC9924943 DOI: 10.1111/acel.13783] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 10/31/2022] [Accepted: 01/05/2023] [Indexed: 01/24/2023] Open
Abstract
Although aging enhances atherosclerosis, we do not know if this occurs via alterations in circulating immune cells, lipid metabolism, vasculature, or adipose tissue. Here, we examined whether aging exerts a direct pro-atherogenic effect on adipose tissue in mice. After demonstrating that aging augmented the inflammatory profile of visceral but not subcutaneous adipose tissue, we transplanted visceral fat from young or aged mice onto the right carotid artery of Ldlr-/- recipients. Aged fat transplants not only increased atherosclerotic plaque size with increased macrophage numbers in the adjacent carotid artery, but also in distal vascular territories, indicating that aging of the adipose tissue enhances atherosclerosis via secreted factors. By depleting macrophages from the visceral fat, we identified that adipose tissue macrophages are major contributors of the secreted factors. To identify these inflammatory factors, we found that aged fat transplants secreted increased levels of the inflammatory mediators TNFα, CXCL2, and CCL2, which synergized to promote monocyte chemotaxis. Importantly, the combined blockade of these inflammatory mediators impeded the ability of aged fat transplants to enhance atherosclerosis. In conclusion, our study reveals that aging enhances atherosclerosis via increased inflammation of visceral fat. Our study suggests that future therapies targeting the visceral fat may reduce atherosclerosis disease burden in the expanding older population.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Diana Farris
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Paola Ariza
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Smriti Moorjani
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Mita Varghese
- Department of Pediatrics, Division of EndocrinologyUniversity of MichiganAnn ArborMichiganUSA
| | - Muriel Blin
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Judy Chen
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
- Graduate Program in ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| | - Daniel Tyrrell
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Min Zhang
- Department of BiostatisticsUniversity of MichiganAnn ArborMichiganUSA
| | - Kanakadurga Singer
- Department of Pediatrics, Division of EndocrinologyUniversity of MichiganAnn ArborMichiganUSA
- Graduate Program in ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| | - Morgan Salmon
- Department of Cardiac SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Daniel R. Goldstein
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
- Graduate Program in ImmunologyUniversity of MichiganAnn ArborMichiganUSA
- Department of Microbiology and ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
19
|
Chen W, Meng F, Zeng X, Cao X, Bu G, Du X, Yu G, Kong F, Li Y, Gan T, Han X. Mechanic Insight into the Distinct and Common Roles of Ovariectomy Versus Adrenalectomy on Adipose Tissue Remodeling in Female Mice. Int J Mol Sci 2023; 24:ijms24032308. [PMID: 36768630 PMCID: PMC9916485 DOI: 10.3390/ijms24032308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Dysfunctions of the ovaries and adrenal glands are both evidenced to cause aberrant adipose tissue (AT) remodeling and resultant metabolic disorders, but their distinct and common roles are poorly understood. In this study, through biochemical, histological and RNA-seq analyses, we comprehensively explored the mechanisms underpinning subcutaneous (SAT) and visceral adipose tissue (VAT) remodeling, in response to ovariectomy (OVX) versus adrenalectomy (ADX) in female mice. OVX promoted adipocyte differentiation and fat accumulation in both SAT and VAT, by potentiating the Pparg signaling, while ADX universally prevented the cell proliferation and extracellular matrix organization in both SAT and VAT, likely by inactivating the Nr3c1 signaling, thus causing lipoatrophy in females. ADX, but not OVX, exerted great effects on the intrinsic difference between SAT and VAT. Specifically, ADX reversed a large cluster of genes differentially expressed between SAT and VAT, by activating 12 key transcription factors, and thereby caused senescent cell accumulation, massive B cell infiltration and the development of selective inflammatory response in SAT. Commonly, both OVX and ADX enhance circadian rhythmicity in VAT, and impair cell proliferation, neurogenesis, tissue morphogenesis, as well as extracellular matrix organization in SAT, thus causing dysfunction of adipose tissues and concomitant metabolic disorders.
Collapse
|
20
|
The Contribution of Tumor Derived Exosomes to Cancer Cachexia. Cells 2023; 12:cells12020292. [PMID: 36672227 PMCID: PMC9856599 DOI: 10.3390/cells12020292] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023] Open
Abstract
Cancer cachexia is defined as unintentional weight loss secondary to neoplasia and is associated with poor prognosis and outcomes. Cancer cachexia associated weight loss affects both lean tissue (i.e., skeletal muscle) and adipose tissue. Exosomes are extracellular vesicles that originate from multivesicular bodies that contain intentionally loaded biomolecular cargo. Exosome cargo includes proteins, lipids, mitochondrial components, and nucleic acids. The cargo carried in exosomes is thought to alter cell signaling when it enters into recipient cells. Virtually every cell type secretes exosomes and exosomes are known to be present in nearly every biofluid. Exosomes alter muscle and adipose tissue metabolism and biological processes, including macrophage polarization and apoptosis which contribute to the development of the cachexia phenotype. This has led to an interest in the role of tumor cell derived exosomes and their potential role as biomarkers of cancer cell development as well as their contribution to cachexia and disease progression. In this review, we highlight published findings that have studied the effects of tumor derived exosomes (and extracellular vesicles) and their cargo on the progression of cancer cachexia. We will focus on the direct effects of tumor derived exosomes and their cellular cross talk on skeletal muscle and adipose tissue, the primary sites of weight loss due to cancer cachexia.
Collapse
|
21
|
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
|
22
|
Jiang S, Lin J, Zhang Q, Liao Y, Lu F, Cai J. The fates of different types of adipose tissue after transplantation in mice. FASEB J 2022; 36:e22510. [PMID: 36004579 DOI: 10.1096/fj.202200408r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/27/2022] [Accepted: 08/09/2022] [Indexed: 11/11/2022]
Abstract
Fat grafting is one of the most commonly applied procedure for soft-tissue repair. However, it remains unclear whether the type of adipose tissue would have any effects on fat graft survival. The present study aimed to determine fates of fat grafting of three different types of fat tissue. In this study, mice were randomly divided into three groups, white adipose tissue (WAT) group, beige adipose tissue (beige AT) group and brown adipose tissue (BAT) group. Before transplantation, donor mice were injected with rosiglitazone or phosphate-buffered saline (PBS). The WAT and BAT were obtained from PBS-treated mice while beige AT was obtained from the rosiglitazone-treated mice. Three types of fat tissue (150 mg each) were transplanted in three groups, respectively, and harvested at 2, 4 or 12 weeks. The BAT and beige AT contained smaller adipocytes and expressed higher level of uncoupling protein-1 gene. The retention rate of the transplanted fat was significantly higher for beige than for white fat, but was significantly lower for brown than for white fat. Transplanted brown fat was characterized by upregulated inflammation and high endoplasmic reticulum stress. By contrast, fat grafts in beige AT group showed the best adipogenic capacity, moderate inflammation level and superior angiongenesis. In vitro, under hypoxic condition, fewer apoptotic cells were found in beige adipocyte group than that in brown and white adipocyte group. Conditioned medium from brown adipocytes induced M1 polarization of RAW 264.7 macrophages while that from beige adipocytes effectively promoted M2 polarization. Therefore, we suggest that beige AT provides a new potential choice for fat grafting because of low inflammation and superior survival but BAT might not be ideal for fat grafting due to its poor survival.
Collapse
Affiliation(s)
- Shenglu Jiang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China.,Department of Basic Medical Sciences, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, P. R. China
| | - Jiayan Lin
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Qian Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Junrong Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
23
|
Ultrasound Imaging of the Superficial Fascia in the Upper Limb: Arm and Forearm. Diagnostics (Basel) 2022; 12:diagnostics12081884. [PMID: 36010234 PMCID: PMC9406830 DOI: 10.3390/diagnostics12081884] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/26/2022] [Accepted: 07/31/2022] [Indexed: 12/29/2022] Open
Abstract
The superficial fascia has received much attention in recent years due to its important role of compartmentalizing the subcutaneous tissue. Ultrasound (US) imaging, owing to its high definition, provides the possibility of better visualizing and measuring its thickness. The aim of this study was to measure and compare, with US imaging, the thickness of superficial fascia in the arm and forearm in different regions/levels. An observational study has been performed using US imaging to measure superficial fascia thickness in the anterior and posterior regions at different levels in a sample of 30 healthy volunteers. The results for superficial fascia thickness revealed statistically significant differences (p < 0.0001) in the arm between the anterior and posterior regions; in terms of forearm, some statistically significant differences were found between regions/levels. However, in the posterior region/levels of the arm, the superficial fascia was thicker (0.53 ± 0.10 mm) than in the forearm (0.41 ± 0.10 mm); regarding the anterior regions/levels, the superficial fascia of the arm (0.40 ± 0.10 mm) was not statistically different than the forearm (0.40 ± 0.12 mm). In addition, the intra-rater reliability was good (ICC2,k: 0.88). US helps to visualize and assess the superficial fascia inside the subcutaneous tissue, improving the diagnosis of fascial dysfunction, and one of the Us parameters to reliably assess is the thickness in different regions and levels.
Collapse
|
24
|
Córdoba-Sosa G, Nicolás-Toledo L, Cervantes-Rodríguez M, Xelhuantzi-Arreguin N, Arteaga-Castañeda MDL, Zambrano E, Cuevas-Romero E, Rodríguez-Antolín J. Maternal and Offspring Sugar Consumption Increases Perigonadal Adipose Tissue Hypertrophy and Negatively Affects the Testis Histological Organization in Adult Rats. Front Cell Dev Biol 2022; 10:893099. [PMID: 35784458 PMCID: PMC9247188 DOI: 10.3389/fcell.2022.893099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/05/2022] [Indexed: 11/21/2022] Open
Abstract
Sugar intake has been associated with the development of male reproductive pathologies because of the increase and dysfunction in different adipose tissue depots. The establishment of these dysfunctions in the early stages of development is unknown. We evaluated the effect of maternal (pregnancy and lactation) and male offspring (from weaning to adulthood) consumption of 5% sucrose on perigonadal adipose tissue (PAT) and testis in adulthood. Moreover, two rat groups were compared, both including pregnant and lactating females: Control (C—drinking tap water) and sugar (S—consuming 5% sucrose solution). From weaning to adulthood with male offspring, four subgroups were formed: Control Mother → Control and Sugar offspring (CC, CS) and Sugar Mother → Control and Sugar offspring (SC, SS). At 120 postnatal days, the testes and PAT were collected and morphologically described. Furthermore, we quantified the number and cross-sectional area of perigonadal adipocytes and their distribution. We found that the males from SC and SS groups showed high PAT weight (p < 0.005), a high number (p < 0.05), and a relative frequency of large adipocytes (p < 0.05), establishing these results during gestational and lactation stages, and enhancing in adulthood since postnatal diet and its interaction. More macrophages, mast cells, and Leydig cells were observed in the interstitial space of the testis for the CS, SC, and SS groups, concluding that consumption of a high-carbohydrate maternal diet, program hypertrophy processes in adult PAT, developing and enhancing with sugar consumption during postnatal life. Furthermore, they are associated with inflammatory processes within the interstitial space of the testis.
Collapse
Affiliation(s)
- Gabriela Córdoba-Sosa
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Leticia Nicolás-Toledo
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | | | - Nicté Xelhuantzi-Arreguin
- Licenciatura en Medicina, Universidad Popular del Estado de Tlaxcala, Tlaxcala, Mexico
- Licenciatura en Enfermería y Obstetricia, Facultad de Ciencias de la Salud, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | | | - Elena Zambrano
- Departamento de Biología Reproductiva, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Estela Cuevas-Romero
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Jorge Rodríguez-Antolín
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
- *Correspondence: Jorge Rodríguez-Antolín,
| |
Collapse
|
25
|
Silvério R, Barth R, Heimann AS, Reckziegel P, dos Santos GJ, Romero-Zerbo SY, Bermúdez-Silva FJ, Rafacho A, Ferro ES. Pep19 Has a Positive Effect on Insulin Sensitivity and Ameliorates Both Hepatic and Adipose Tissue Phenotype of Diet-Induced Obese Mice. Int J Mol Sci 2022; 23:ijms23084082. [PMID: 35456900 PMCID: PMC9030859 DOI: 10.3390/ijms23084082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 12/10/2022] Open
Abstract
Peptide DIIADDEPLT (Pep19) has been previously suggested to improve metabolic parameters, without adverse central nervous system effects, in a murine model of diet-induced obesity. Here, we aimed to further evaluate whether Pep19 oral administration has anti-obesogenic effects, in a well-established high-fat diet-induced obesity model. Male Swiss mice, fed either a standard diet (SD) or high-fat diet (HFD), were orally administrated for 30 consecutive days, once a day, with saline vehicle or Pep19 (1 mg/kg). Next, several metabolic, morphological, and behavioral parameters were evaluated. Oral administration of Pep19 attenuated HFD body-weight gain, reduced in approximately 40% the absolute mass of the endocrine pancreas, and improved the relationship between circulating insulin and peripheral insulin sensitivity. Pep19 treatment of HFD-fed mice attenuated liver inflammation, hepatic fat distribution and accumulation, and lowered plasma alanine aminotransferase activity. The inguinal fat depot from the SD group treated with Pep19 showed multilocular brown-fat-like cells and increased mRNA expression of uncoupling protein 1 (UCP1), suggesting browning on inguinal white adipose cells. Morphological analysis of brown adipose tissue (BAT) from HFD mice showed the presence of larger white-like unilocular cells, compared to BAT from SD, Pep19-treated SD or HFD mice. Pep19 treatment produced no alterations in mice behavior. Oral administration of Pep19 ameliorates some metabolic traits altered by diet-induced obesity in a Swiss mice model.
Collapse
Affiliation(s)
- Renata Silvério
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, Brazil;
- Laboratory of Investigation in Chronic Diseases, Department of Physiological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, Brazil; (R.B.); (G.J.d.S.)
| | - Robson Barth
- Laboratory of Investigation in Chronic Diseases, Department of Physiological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, Brazil; (R.B.); (G.J.d.S.)
- Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, Brazil
| | - Andrea S. Heimann
- Proteimax BioTechnology Israel LTD, 4 Duvdevan Street, Pardes Hana, Haifa 3708973, Israel;
| | - Patrícia Reckziegel
- Department of Pharmacology, Biomedical Science Institute, University of São Paulo (USP), São Paulo 05508-000, Brazil;
| | - Gustavo J. dos Santos
- Laboratory of Investigation in Chronic Diseases, Department of Physiological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, Brazil; (R.B.); (G.J.d.S.)
- Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, Brazil
| | - Silvana Y. Romero-Zerbo
- Instituto de Investigación Biomédica de Málaga-IBIMA, UGC Endocrinología y Nutrición Hospital Regional Universitario de Málaga, Universidad de Málaga, 29009 Málaga, Spain; (S.Y.R.-Z.); (F.J.B.-S.)
- Biomedical Research Center for Diabetes and Associated Metabolic Diseases (CIBERDEM), 28029 Madrid, Spain
| | - Francisco J. Bermúdez-Silva
- Instituto de Investigación Biomédica de Málaga-IBIMA, UGC Endocrinología y Nutrición Hospital Regional Universitario de Málaga, Universidad de Málaga, 29009 Málaga, Spain; (S.Y.R.-Z.); (F.J.B.-S.)
- Biomedical Research Center for Diabetes and Associated Metabolic Diseases (CIBERDEM), 28029 Madrid, Spain
| | - Alex Rafacho
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, Brazil;
- Laboratory of Investigation in Chronic Diseases, Department of Physiological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, Brazil; (R.B.); (G.J.d.S.)
- Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, Brazil
- Correspondence: (A.R.); (E.S.F.)
| | - Emer S. Ferro
- Department of Pharmacology, Biomedical Science Institute, University of São Paulo (USP), São Paulo 05508-000, Brazil;
- Correspondence: (A.R.); (E.S.F.)
| |
Collapse
|
26
|
Romero AR, Mu A, Ayres JS. Adipose triglyceride lipase mediates lipolysis and lipid mobilization in response to iron-mediated negative energy balance. iScience 2022; 25:103941. [PMID: 35265813 PMCID: PMC8899412 DOI: 10.1016/j.isci.2022.103941] [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: 09/13/2021] [Revised: 12/23/2021] [Accepted: 02/14/2022] [Indexed: 11/09/2022] Open
Abstract
Maintenance of energy balance is essential for overall organismal health. Mammals have evolved complex regulatory mechanisms that control energy intake and expenditure. Traditionally, studies have focused on understanding the role of macronutrient physiology in energy balance. In the present study, we examined the role of the essential micronutrient iron in regulating energy balance. We found that a short course of dietary iron caused a negative energy balance resulting in a severe whole body wasting phenotype. This disruption in energy balance was because of impaired intestinal nutrient absorption. In response to dietary iron-induced negative energy balance, adipose triglyceride lipase (ATGL) was necessary for wasting of subcutaneous white adipose tissue and lipid mobilization. Fat-specific ATGL deficiency protected mice from fat wasting, but caused a severe cachectic response in mice when fed iron. Our work reveals a mechanism for micronutrient control of lipolysis that is necessary for regulating mammalian energy balance.
Collapse
Affiliation(s)
- Alicia R. Romero
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Gene Expression Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Division of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Andre Mu
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Gene Expression Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Janelle S. Ayres
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Gene Expression Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Corresponding author
| |
Collapse
|
27
|
Role of Distinct Fat Depots in Metabolic Regulation and Pathological Implications. Rev Physiol Biochem Pharmacol 2022; 186:135-176. [PMID: 35915363 DOI: 10.1007/112_2022_73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
People suffering from obesity and associated metabolic disorders including diabetes are increasing exponentially around the world. Adipose tissue (AT) distribution and alteration in their biochemical properties play a major role in the pathogenesis of these diseases. Emerging evidence suggests that AT heterogeneity and depot-specific physiological changes are vital in the development of insulin resistance in peripheral tissues like muscle and liver. Classically, AT depots are classified into white adipose tissue (WAT) and brown adipose tissue (BAT); WAT is the site of fatty acid storage, while BAT is a dedicated organ of metabolic heat production. The discovery of beige adipocyte clusters in WAT depots indicates AT heterogeneity has a more central role than hither to ascribed. Therefore, we have discussed in detail the current state of understanding on cellular and molecular origin of different AT depots and their relevance toward physiological metabolic homeostasis. A major focus is to highlight the correlation between altered WAT distribution in the body and metabolic pathogenesis in animal models and humans. We have also underscored the disparity in the molecular (including signaling) changes in various WAT tissues during diabetic pathogenesis. Exercise-mediated beneficial alteration in WAT physiology/distribution that protects against metabolic disorders is evolving. Here we have discussed the depot-specific biochemical adjustments induced by different forms of exercise. A detailed understanding of the molecular details of inter-organ crosstalk via substrate utilization/storage and signaling through chemokines provide strategies to target selected WAT depots to pharmacologically mimic the benefits of exercise countering metabolic diseases including diabetes.
Collapse
|
28
|
Le Lay S, Magré J, Prieur X. Not Enough Fat: Mouse Models of Inherited Lipodystrophy. Front Endocrinol (Lausanne) 2022; 13:785819. [PMID: 35250856 PMCID: PMC8895270 DOI: 10.3389/fendo.2022.785819] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Lipodystrophies belong to the heterogenous group of syndromes in which the primary defect is a generalized or partial absence of adipose tissue, which may be congenital or acquired in origin. Lipodystrophy should be considered in patients manifesting the combination of insulin resistance (with or without overt diabetes), dyslipidemia and fatty liver. Lipodystrophies are classified according to the etiology of the disease (genetic or acquired) and to the anatomical distribution of adipose tissue (generalized or partial). The mechanism of adipose tissue loss is specific to each syndrome, depending on the biological function of the mutated gene. Mice models, together with cellular studies have permitted clarification of the mechanisms by which human mutations deeply compromise adipocyte homeostasis. In addition, rodent models have proven to be crucial in deciphering the cardiometabolic consequences of the lack of adipose tissue such as NAFLD, muscle insulin resistance and cardiomyopathy. More precisely, tissue-specific transgenic and knockout mice have brought new tools to distinguish phenotypic traits that are the consequences of lipodystrophy from those that are cell-autonomous. In this review, we discuss the mice models of lipodystrophy including those of inherited human syndromes of generalized and partial lipodystrophy. We present how these models have demonstrated the central role of white adipose tissue in energetic homeostasis in general, including insulin sensitivity and lipid handling in particular. We underscore the differences reported with the human phenotype and discuss the limit of rodent models in recapitulating adipose tissue primary default. Finally, we present how these mice models have highlighted the function of the causative-genes and brought new insights into the pathophysiology of the cardiometabolic complications associated with lipodystrophy.
Collapse
Affiliation(s)
- Soazig Le Lay
- Nantes Université, CNRS, INSERM, l’institut du thorax, Nantes, France
- Univ Angers, SFR ICAT, Angers, France
| | - Jocelyne Magré
- Nantes Université, CNRS, INSERM, l’institut du thorax, Nantes, France
| | - Xavier Prieur
- Nantes Université, CNRS, INSERM, l’institut du thorax, Nantes, France
- *Correspondence: Xavier Prieur,
| |
Collapse
|
29
|
Ko SH, Jung Y. Energy Metabolism Changes and Dysregulated Lipid Metabolism in Postmenopausal Women. Nutrients 2021; 13:nu13124556. [PMID: 34960109 PMCID: PMC8704126 DOI: 10.3390/nu13124556] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
Aging women experience hormonal changes, such as decreased estrogen and increased circulating androgen, due to natural or surgical menopause. These hormonal changes make postmenopausal women vulnerable to body composition changes, muscle loss, and abdominal obesity; with a sedentary lifestyle, these changes affect overall energy expenditure and basal metabolic rate. In addition, fat redistribution due to hormonal changes leads to changes in body shape. In particular, increased bone marrow-derived adipocytes due to estrogen loss contribute to increased visceral fat in postmenopausal women. Enhanced visceral fat lipolysis by adipose tissue lipoprotein lipase triggers the production of excessive free fatty acids, causing insulin resistance and metabolic diseases. Because genes involved in β-oxidation are downregulated by estradiol loss, excess free fatty acids produced by lipolysis of visceral fat cannot be used appropriately as an energy source through β-oxidation. Moreover, aged women show increased adipogenesis due to upregulated expression of genes related to fat accumulation. As a result, the catabolism of ATP production associated with β-oxidation decreases, and metabolism associated with lipid synthesis increases. This review describes the changes in energy metabolism and lipid metabolic abnormalities that are the background of weight gain in postmenopausal women.
Collapse
Affiliation(s)
- Seong-Hee Ko
- Department of Microbiology, College of Medicine, Gachon University, Incheon 21999, Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea
| | - YunJae Jung
- Department of Microbiology, College of Medicine, Gachon University, Incheon 21999, Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea
- Correspondence: ; Tel.: +82-32-899-6415
| |
Collapse
|
30
|
Knuth CM, Auger C, Chi L, Barayan D, Abdullahi A, Jeschke MG. Thermal Stress Induces Long-Term Remodeling of Adipose Tissue and Is Associated with Systemic Dysfunction. Shock 2021; 56:744-754. [PMID: 33534398 PMCID: PMC8316494 DOI: 10.1097/shk.0000000000001743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
ABSTRACT Severe burns are characterized by the magnitude and duration of the hypermetabolic response thereafter, and demarcated by the loss of lean body mass and catabolism of fat stores. The aim of the present study was to delineate the temporal and location-specific physiological changes to adipose depots and downstream consequences post-burn in a murine model of thermal injury. C57BL/6 mice were subjected to a 30% total body surface area burn and body mass, food intake, and tissue mass were monitored for various time points up until 60 days postinjury. Mitochondrial respirometry was performed using a Seahorse XF96 analyzer. Lipolytic markers and browning markers were analyzed via Western blotting and histology. A severe burn results in a futile cycle of lipolysis and white adipose tissue (WAT) browning, the sequelae of which include fat catabolism, hepatomegaly, and loss of body mass despite increased food intake. A dynamic remodeling of epididymal WAT was observed with acute and chronic increases in lipolysis. Moreover, we demonstrate that pathological browning of inguinal WAT persists up to 60 days post-burn, highlighting the magnitude of the β-adrenergic response to thermal injury. Our data suggests that adipose depots have a heterogeneous response to burns and that therapeutic interventions targeting these physiological changes can improve outcomes. These data may also have implications for treating catabolic conditions such as cancer cachexia as well as developing treatments for obesity and type II diabetes.
Collapse
Affiliation(s)
- Carly M. Knuth
- Institute of Medical Science, University of Toronto, Canada
| | | | - Leon Chi
- Sunnybrook Research Institute, Toronto, Canada
| | - Dalia Barayan
- Institute of Medical Science, University of Toronto, Canada
| | | | - Marc G. Jeschke
- Institute of Medical Science, University of Toronto, Canada
- Department of Immunology, University of Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
- Department of Surgery, Division of Plastic Surgery, University of Toronto, Canada
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| |
Collapse
|
31
|
Sánchez YL, Yepes-Calderón M, Valbuena L, Milán AF, Trillos-Almanza MC, Granados S, Peña M, Estrada-Castrillón M, Aristizábal JC, Narvez-Sanchez R, Gallo-Villegas J, Calderón JC. Musclin Is Related to Insulin Resistance and Body Composition, but Not to Body Mass Index or Cardiorespiratory Capacity in Adults. Endocrinol Metab (Seoul) 2021; 36:1055-1068. [PMID: 34674511 PMCID: PMC8566119 DOI: 10.3803/enm.2021.1104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/27/2021] [Accepted: 08/24/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND We studied whether musclin function in humans is related to glycemic control, body composition, and cardiorespiratory capacity. METHODS A cross-sectional study was performed in sedentary adults with or without metabolic syndrome (MS). Serum musclin was measured by enzyme-linked immunosorbent assay. Insulin resistance (IR) was evaluated by the homeostatic model assessment (HOMA-IR). Body composition was determined by dual-energy X-ray absorptiometry and muscle composition by measuring carnosine in the thigh, a surrogate of fiber types, through proton magnetic resonance spectroscopy. Cardiorespiratory capacity was assessed through direct ergospirometry. RESULTS The control (n=29) and MS (n=61) groups were comparable in age (51.5±6.5 years old vs. 50.7±6.1 years old), sex (72.4% vs. 70.5% women), total lean mass (58.5%±7.4% vs. 57.3%±6.8%), and peak oxygen consumption (VO2peak) (31.0±5.8 mL O2./kg.min vs. 29.2±6.3 mL O2/kg.min). Individuals with MS had higher body mass index (BMI) (30.6±4.0 kg/m2 vs. 27.4± 3.6 kg/m2), HOMA-IR (3.5 [95% confidence interval, CI, 2.9 to 4.6] vs. 1.7 [95% CI, 1.1 to 2.0]), and musclin (206.7 pg/mL [95% CI, 122.7 to 387.8] vs. 111.1 pg/mL [95% CI, 63.2 to 218.5]) values than controls (P˂0.05). Musclin showed a significant relationship with HOMA-IR (β=0.23; 95% CI, 0.12 to 0.33; P˂0.01), but not with VO2peak, in multiple linear regression models adjusted for age, sex, fat mass, lean mass, and physical activity. Musclin was significantly associated with insulin, glycemia, visceral fat, and regional muscle mass, but not with BMI, VCO2peak, maximum heart rate, maximum time of work, or carnosine. CONCLUSION In humans, musclin positively correlates with insulinemia, IR, and a body composition profile with high visceral adiposity and lean mass, but low body fat percentage. Musclin is not related to BMI or cardiorespiratory capacity.
Collapse
Affiliation(s)
- Yeliana L. Sánchez
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
| | - Manuela Yepes-Calderón
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
| | - Luis Valbuena
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
- Indeportes Antioquia, Medellin,
Colombia
| | - Andrés F. Milán
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
| | - María C. Trillos-Almanza
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
| | - Sergio Granados
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
| | - Miguel Peña
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
| | | | - Juan C. Aristizábal
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
| | - Raúl Narvez-Sanchez
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
| | - Jaime Gallo-Villegas
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
- Sports Medicine Postgraduate Program, and GRINMADE Research Group, SICOR Center, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
| | - Juan C. Calderón
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellin,
Colombia
| |
Collapse
|
32
|
Lepanto P, Levin-Ferreyra F, Koziol U, Malacrida L, Badano JL. Insights into in vivo adipocyte differentiation through cell-specific labeling in zebrafish. Biol Open 2021; 10:271875. [PMID: 34409430 PMCID: PMC8443861 DOI: 10.1242/bio.058734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/10/2021] [Indexed: 01/04/2023] Open
Abstract
White adipose tissue hyperplasia has been shown to be crucial for handling excess energy in healthy ways. Though adipogenesis mechanisms have been underscored in vitro, we lack information on how tissue and systemic factors influence the differentiation of new adipocytes. While this could be studied in zebrafish, adipocyte identification currently relies on neutral lipid labeling, thus precluding access to cells in early stages of differentiation. Here we report the generation and analysis of a zebrafish line with the transgene fabp4a(-2.7):EGFPcaax. In vivo confocal microscopy of the pancreatic and abdominal visceral depots of transgenic larvae, revealed the presence of labeled mature adipocytes as well as immature cells in earlier stages of differentiation. Through co-labeling for blood vessels, we observed a close interaction of differentiating adipocytes with endothelial cells through cell protrusions. Finally, we implemented hyperspectral imaging and spectral phasor analysis in Nile Red-labeled transgenic larvae and revealed the lipid metabolic transition towards neutral lipid accumulation of differentiating adipocytes. Altogether our work presents the characterization of a novel adipocyte-specific label in zebrafish and uncovers previously unknown aspects of in vivo adipogenesis. This article has an associated First Person interview with the first author of the paper. Summary: Analysis of the differentiation of adipocytes in vivo through cell-specific labeling in zebrafish, revealed their early interaction with blood vessels as well as early lipid metabolic changes.
Collapse
Affiliation(s)
- Paola Lepanto
- Human Molecular Genetics Lab, Institut Pasteur de Montevideo, Montevideo, Mataojo 2020, CP11400, Uruguay
| | - Florencia Levin-Ferreyra
- Human Molecular Genetics Lab, Institut Pasteur de Montevideo, Montevideo, Mataojo 2020, CP11400, Uruguay
| | - Uriel Koziol
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Igua 4225, CP11400, Uruguay
| | - Leonel Malacrida
- Advanced Bioimaging Unit, Institut Pasteur de Montevideo and Universidad de la República, Montevideo, Mataojo 2020, CP11400, Uruguay.,Departamento de Fisiopatología, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Av. Italia s/n, CP11600, Uruguay
| | - José L Badano
- Human Molecular Genetics Lab, Institut Pasteur de Montevideo, Montevideo, Mataojo 2020, CP11400, Uruguay
| |
Collapse
|
33
|
Adipose-Derived Exosomes as Possible Players in the Development of Insulin Resistance. Int J Mol Sci 2021; 22:ijms22147427. [PMID: 34299048 PMCID: PMC8304687 DOI: 10.3390/ijms22147427] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/25/2022] Open
Abstract
Adipose tissue (AT) is an endocrine organ involved in the management of energy metabolism via secretion of adipokines, hormones, and recently described secretory microvesicles, i.e., exosomes. Exosomes are rich in possible biologically active factors such as proteins, lipids, and RNA. The secretory function of adipose tissue is affected by pathological processes. One of the most important of these is obesity, which triggers adipose tissue inflammation and adversely affects the release of beneficial adipokines. Both processes may lead to further AT dysfunction, contributing to changes in whole-body metabolism and, subsequently, to insulin resistance. According to recent data, changes within the production, release, and content of exosomes produced by AT may be essential to understand the role of adipose tissue in the development of metabolic disorders. In this review, we summarize actual knowledge about the possible role of AT-derived exosomes in the development of insulin resistance, highlighting methodological challenges and potential gains resulting from exosome studies.
Collapse
|
34
|
Ong WK, Chakraborty S, Sugii S. Adipose Tissue: Understanding the Heterogeneity of Stem Cells for Regenerative Medicine. Biomolecules 2021; 11:biom11070918. [PMID: 34206204 PMCID: PMC8301750 DOI: 10.3390/biom11070918] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stem cells (ASCs) have been increasingly used as a versatile source of mesenchymal stem cells (MSCs) for diverse clinical investigations. However, their applications often become complicated due to heterogeneity arising from various factors. Cellular heterogeneity can occur due to: (i) nomenclature and criteria for definition; (ii) adipose tissue depots (e.g., subcutaneous fat, visceral fat) from which ASCs are isolated; (iii) donor and inter-subject variation (age, body mass index, gender, and disease state); (iv) species difference; and (v) study design (in vivo versus in vitro) and tools used (e.g., antibody isolation and culture conditions). There are also actual differences in resident cell types that exhibit ASC/MSC characteristics. Multilineage-differentiating stress-enduring (Muse) cells and dedifferentiated fat (DFAT) cells have been reported as an alternative or derivative source of ASCs for application in regenerative medicine. In this review, we discuss these factors that contribute to the heterogeneity of human ASCs in detail, and what should be taken into consideration for overcoming challenges associated with such heterogeneity in the clinical use of ASCs. Attempts to understand, define, and standardize cellular heterogeneity are important in supporting therapeutic strategies and regulatory considerations for the use of ASCs.
Collapse
Affiliation(s)
- Wee Kiat Ong
- School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Selangor, Malaysia
- Correspondence: (W.K.O.); (S.S.)
| | - Smarajit Chakraborty
- Institute of Bioengineering and Bioimaging (IBB), A*STAR, 31 Biopolis Way, Singapore 138669, Singapore;
| | - Shigeki Sugii
- Institute of Bioengineering and Bioimaging (IBB), A*STAR, 31 Biopolis Way, Singapore 138669, Singapore;
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Correspondence: (W.K.O.); (S.S.)
| |
Collapse
|
35
|
Morigny P, Boucher J, Arner P, Langin D. Lipid and glucose metabolism in white adipocytes: pathways, dysfunction and therapeutics. Nat Rev Endocrinol 2021; 17:276-295. [PMID: 33627836 DOI: 10.1038/s41574-021-00471-8] [Citation(s) in RCA: 240] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 12/14/2022]
Abstract
In mammals, the white adipocyte is a cell type that is specialized for storage of energy (in the form of triacylglycerols) and for energy mobilization (as fatty acids). White adipocyte metabolism confers an essential role to adipose tissue in whole-body homeostasis. Dysfunction in white adipocyte metabolism is a cardinal event in the development of insulin resistance and associated disorders. This Review focuses on our current understanding of lipid and glucose metabolic pathways in the white adipocyte. We survey recent advances in humans on the importance of adipocyte hypertrophy and on the in vivo turnover of adipocytes and stored lipids. At the molecular level, the identification of novel regulators and of the interplay between metabolic pathways explains the fine-tuning between the anabolic and catabolic fates of fatty acids and glucose in different physiological states. We also examine the metabolic alterations involved in the genesis of obesity-associated metabolic disorders, lipodystrophic states, cancers and cancer-associated cachexia. New challenges include defining the heterogeneity of white adipocytes in different anatomical locations throughout the lifespan and investigating the importance of rhythmic processes. Targeting white fat metabolism offers opportunities for improved patient stratification and a wide, yet unexploited, range of therapeutic opportunities.
Collapse
Affiliation(s)
- Pauline Morigny
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1297, Toulouse, France
- University of Toulouse, Paul Sabatier University, I2MC, UMR1297, Toulouse, France
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jeremie Boucher
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- The Lundberg Laboratory for Diabetes Research, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Peter Arner
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Dominique Langin
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1297, Toulouse, France.
- University of Toulouse, Paul Sabatier University, I2MC, UMR1297, Toulouse, France.
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague and Paul Sabatier University, Toulouse, France.
- Toulouse University Hospitals, Laboratory of Clinical Biochemistry, Toulouse, France.
| |
Collapse
|
36
|
Mathiesen A, Hamilton T, Carter N, Brown M, McPheat W, Dobrian A. Endothelial Extracellular Vesicles: From Keepers of Health to Messengers of Disease. Int J Mol Sci 2021; 22:ijms22094640. [PMID: 33924982 PMCID: PMC8125116 DOI: 10.3390/ijms22094640] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 02/07/2023] Open
Abstract
Endothelium has a rich vesicular network that allows the exchange of macromolecules between blood and parenchymal cells. This feature of endothelial cells, along with their polarized secretory machinery, makes them the second major contributor, after platelets, to the particulate secretome in circulation. Extracellular vesicles (EVs) produced by the endothelial cells mirror the remarkable molecular heterogeneity of their parent cells. Cargo molecules carried by EVs were shown to contribute to the physiological functions of endothelium and may support the plasticity and adaptation of endothelial cells in a paracrine manner. Endothelium-derived vesicles can also contribute to the pathogenesis of cardiovascular disease or can serve as prognostic or diagnostic biomarkers. Finally, endothelium-derived EVs can be used as therapeutic tools to target endothelium for drug delivery or target stromal cells via the endothelial cells. In this review we revisit the recent evidence on the heterogeneity and plasticity of endothelial cells and their EVs. We discuss the role of endothelial EVs in the maintenance of vascular homeostasis along with their contributions to endothelial adaptation and dysfunction. Finally, we evaluate the potential of endothelial EVs as disease biomarkers and their leverage as therapeutic tools.
Collapse
|
37
|
Dou J, Puttabyatappa M, Padmanabhan V, Bakulski KM. Developmental programming: Adipose depot-specific transcriptional regulation by prenatal testosterone excess in a sheep model of PCOS. Mol Cell Endocrinol 2021; 523:111137. [PMID: 33359827 PMCID: PMC7854529 DOI: 10.1016/j.mce.2020.111137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/16/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023]
Abstract
Prenatal testosterone (T)-treated female sheep manifest adipose depot-specific disruptions in inflammatory/oxidative state, adipocyte differentiation and thermogenic adipocyte distribution. The objective of this study was to identify common and divergent gene pathways underlying prenatal T excess-induced adipose depot-specific disruptions. RNA sequencing and network analyses were undertaken with visceral (VAT), subcutaneous (SAT), epicardiac (ECAT) and perirenal (PRAT) adipose tissues from control and prenatal T-treated (100 mg T propionate twice a week from days 30-90 of gestation) female sheep at 21 months of age. Increased expression of adiposity and inflammation-related genes in VAT and genes that promote differentiation of white adipocytes in SAT were congruous with their metabolic roles with SAT favoring uptake/storage of free fatty acids and triglycerides and VAT favoring higher rate of fatty acid turnover and lipolysis. Selective upregulation of cardiac muscle and renoprotection genes in ECAT and PRAT respectively are suggestive of protective paracrine actions. Expression profile in prenatal T-treated sheep paralleled depot-specific dysfunctions with increased proinflammatory genes in VAT, reduced adipocyte differentiation genes in VAT and SAT and increased vascular related gene expression in PRAT. The high expression of genes involved in cardiomyocyte function in ECAT is suggestive of cardioprotective function being maintained to overcome the prenatal T-induced cardiac dysfunction and hypertension. These findings coupled with changes in gene pathways and networks involved in chromatin modification, extracellular matrix, immune and mitochondrial function, and endoplasmic reticulum to Golgi transport suggest that dysregulation in gene expression underlie prenatal T-treatment induced functional differences among adipose depots and manifestation of metabolic dysfunction.
Collapse
Affiliation(s)
- John Dou
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Kelly M Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
38
|
Chemically Defined Xeno- and Serum-Free Cell Culture Medium to Grow Human Adipose Stem Cells. Cells 2021; 10:cells10020466. [PMID: 33671568 PMCID: PMC7926673 DOI: 10.3390/cells10020466] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue is an abundant source of stem cells. However, liposuction cannot yield cell quantities sufficient for direct applications in regenerative medicine. Therefore, the development of GMP-compliant ex vivo expansion protocols is required to ensure the production of a "cell drug" that is safe, reproducible, and cost-effective. Thus, we developed our own basal defined xeno- and serum-free cell culture medium (UrSuppe), specifically formulated to grow human adipose stem cells (hASCs). With this medium, we can directly culture the stromal vascular fraction (SVF) cells in defined cell culture conditions to obtain hASCs. Cells proliferate while remaining undifferentiated, as shown by Flow Cytometry (FACS), Quantitative Reverse Transcription PCR (RT-qPCR) assays, and their secretion products. Using the UrSuppe cell culture medium, maximum cell densities between 0.51 and 0.80 × 105 cells/cm2 (=2.55-4.00 × 105 cells/mL) were obtained. As the expansion of hASCs represents only the first step in a cell therapeutic protocol or further basic research studies, we formulated two chemically defined media to differentiate the expanded hASCs in white or beige/brown adipocytes. These new media could help translate research projects into the clinical application of hASCs and study ex vivo the biology in healthy and dysfunctional states of adipocytes and their precursors. Following the cell culture system developers' practice and obvious reasons related to the formulas' patentability, the defined media's composition will not be disclosed in this study.
Collapse
|
39
|
Misiou A, Garmey JC, Hensien JM, Harmon DB, Osinski V, McSkimming C, Marshall MA, Fischer JW, Grandoch M, McNamara CA. Helix-Loop-Helix Factor Id3 (Inhibitor of Differentiation 3): A Novel Regulator of Hyaluronan-Mediated Adipose Tissue Inflammation. Arterioscler Thromb Vasc Biol 2021; 41:796-807. [PMID: 33380173 PMCID: PMC8105274 DOI: 10.1161/atvbaha.120.315588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The aim of this study was to unravel mechanisms whereby deficiency of the transcription factor Id3 (inhibitor of differentiation 3) leads to metabolic dysfunction in visceral obesity. We investigated the impact of loss of Id3 on hyaluronic acid (HA) production by the 3 HAS isoenzymes (HA synthases; -1, -2, and -3) and on obesity-induced adipose tissue (AT) accumulation of proinflammatory B cells. Approach and Results: Male Id3-/- mice and respective wild-type littermate controls were fed a 60% high-fat diet for 4 weeks. An increase in inflammatory B2 cells was detected in Id3-/- epididymal AT. HA accumulated in epididymal AT of high-fat diet-fed Id3-/- mice and circulating levels of HA were elevated. Has2 mRNA expression was increased in epididymal AT of Id3-/- mice. Luciferase promoter assays showed that Id3 suppressed Has2 promoter activity, while loss of Id3 stimulated Has2 promoter activity. Functionally, HA strongly promoted B2 cell adhesion in the AT and on cultured vascular smooth muscle cells of Id3-/- mice, an effect sensitive to hyaluronidase. CONCLUSIONS Our data demonstrate that loss of Id3 increases Has2 expression in the epididymal AT, thereby promoting HA accumulation. In turn, elevated HA content promotes HA-dependent binding of B2 cells and an increase in the B2 cells in the AT, which contributes to AT inflammation.
Collapse
MESH Headings
- Adipose Tissue/immunology
- Adipose Tissue/metabolism
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cell Adhesion
- Cells, Cultured
- Coculture Techniques
- Diet, High-Fat
- Disease Models, Animal
- Hyaluronan Synthases/genetics
- Hyaluronan Synthases/metabolism
- Hyaluronic Acid/biosynthesis
- Inhibitor of Differentiation Proteins/genetics
- Inhibitor of Differentiation Proteins/metabolism
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/immunology
- Myocytes, Smooth Muscle/metabolism
- Panniculitis/genetics
- Panniculitis/immunology
- Panniculitis/metabolism
- Phenotype
- Signal Transduction
- Up-Regulation
- Mice
Collapse
Affiliation(s)
- Angelina Misiou
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - James C. Garmey
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Jack M. Hensien
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Daniel B. Harmon
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Victoria Osinski
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Chantel McSkimming
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Melissa A. Marshall
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Jens W. Fischer
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Maria Grandoch
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Coleen A. McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
40
|
Abstract
Adipose, or fat, tissue (AT) was once considered an inert tissue that primarily existed to store lipids, and was not historically recognized as an important organ in the regulation and maintenance of health. With the rise of obesity and more rigorous research, AT is now recognized as a highly complex metabolic organ involved in a host of important physiological functions, including glucose homeostasis and a multitude of endocrine capabilities. AT dysfunction has been implicated in several disease states, most notably obesity, metabolic syndrome and type 2 diabetes. The study of AT has provided useful insight in developing strategies to combat these highly prevalent metabolic diseases. This review highlights the major functions of adipose tissue and the consequences that can occur when disruption of these functions leads to systemic metabolic dysfunction.
Collapse
Affiliation(s)
- Innocence Harvey
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Anik Boudreau
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Jacqueline M Stephens
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.,Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
41
|
Diabetogenic diet-induced insulin resistance associates with lipid droplet proteins and adipose tissue secretome, but not with sexual dimorphic adipose tissue fat accumulation in wistar rats. Biochem Biophys Rep 2020; 24:100831. [PMID: 33088930 PMCID: PMC7559855 DOI: 10.1016/j.bbrep.2020.100831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/26/2019] [Accepted: 10/06/2020] [Indexed: 12/25/2022] Open
Abstract
The role of sexual dimorphic adipose tissue fat accumulation in the development of insulin resistance is well known. However, whether vitamin A status and/or its metabolic pathway display any sex- or depot (visceral/subcutaneous)-specific pattern and have a role in sexual dimorphic adipose tissue development and insulin resistance are not completely understood. Therefore, to assess this, 5 weeks old Wistar male and female rats of eight from each sex were provided either control or diabetogenic (high fat, high sucrose) diet for 26 weeks. At the end, consumption of diabetogenic diet increased the visceral fat depots (p < 0.001) in the males and subcutaneous depot (p < 0.05) in the female rats, compared to their sex-matched controls. On the other hand, it caused adipocyte hypertrophy (p < 0.05) of visceral depot (retroperitoneal) in the females and subcutaneous depot of the male rats. Although vitamin A levels displayed sex- and depot-specific increase due to the consumption of diabetogenic diet, the expression of most of its metabolic pathway genes in adipose depots remained unaltered. However, the mRNA levels of some of lipid droplet proteins (perilipins) and adipose tissue secretory proteins (interleukins, lipocalin-2) did display sexual dimorphism. Nonetheless, the long-term feeding of diabetogenic diet impaired the insulin sensitivity, thus affected glucose clearance rate and muscle glucose-uptake in both the sexes of rats. In conclusion, the chronic consumption of diabetogenic diet caused insulin resistance in the male and female rats, but did not corroborate with sexual dimorphic adipose tissue fat accumulation or its vitamin A status. Role of vitamin A and its metabolic pathway on sexual dimorphic fat accumulation and insulin resistance was studied. Consumption of diabetogenic diet caused insulin resistance, but not associated with sexual-dimorphic fat deposition. Vitamin A accumulation displayed a sex- and fat depot-specific pattern without altering its metabolic pathway genes. However, the lipid droplet proteins and secretome of the adipose depots displayed sex- and/or depot-specific pattern.
Collapse
|
42
|
Dumas JF, Brisson L. Interaction between adipose tissue and cancer cells: role for cancer progression. Cancer Metastasis Rev 2020; 40:31-46. [PMID: 33009650 DOI: 10.1007/s10555-020-09934-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/22/2020] [Indexed: 12/20/2022]
Abstract
Environment surrounding tumours are now recognized to play an important role in tumour development and progression. Among the cells found in the tumour environment, adipocytes from adipose tissue establish a vicious cycle with cancer cells to promote cancer survival, proliferation, metastasis and treatment resistance. This cycle is particularly of interest in the context of obesity, which has been found as a cancer risk factor. Cancers cells can reprogram adipocyte physiology leading to an "activated" phenotype characterized by delipidation and secretion of inflammatory adipokines. The adipocyte secretions then influence tumour growth and metastasis which has been mainly attributed to interleukin 6 (IL-6) or leptin but also to the release of fatty acids which are able to change cancer cell metabolism and signalling pathways. The aim of this review is to report recent advances in the understanding of the molecular mechanisms linking adipose tissue with cancer progression in order to propose new therapeutic strategies based on pharmacological or nutritional intervention.
Collapse
Affiliation(s)
- Jean-François Dumas
- Inserm UMR1069, Nutrition, Growth and Cancer, University of Tours, 10 boulevard Tonnellé, 37032, Tours, France
| | - Lucie Brisson
- Inserm UMR1069, Nutrition, Growth and Cancer, University of Tours, 10 boulevard Tonnellé, 37032, Tours, France.
| |
Collapse
|
43
|
Boldarine VT, Pedroso AP, Brandão-Teles C, LoTurco EG, Nascimento CMO, Oyama LM, Bueno AA, Martins-de-Souza D, Ribeiro EB. Ovariectomy modifies lipid metabolism of retroperitoneal white fat in rats: a proteomic approach. Am J Physiol Endocrinol Metab 2020; 319:E427-E437. [PMID: 32663100 DOI: 10.1152/ajpendo.00094.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Menopause is often accompanied by visceral obesity. With the aim of exploring the consequences of ovarian failure on visceral fat, we evaluated the effects of ovariectomy and estrogen replacement on the proteome/phosphoproteome and on the fatty acid profile of the retroperitoneal adipose depot (RAT) of rats. Eighteen 3-mo-old female Wistar rats were either ovariectomized or sham operated and fed with standard chow for 3 mo. A subgroup of ovariectomized rats received estradiol replacement. RAT samples were analyzed with data-independent acquisitions LC-MS/MS, and pathway analysis was performed with the differentially expressed/phosphorylated proteins. RAT lipid profile was analyzed by gas chromatography. Ovariectomy induced high adiposity and insulin resistance and promoted alterations in protein expression and phosphorylation. Pathway analysis showed that five pathways were significantly affected by ovariectomy, namely, metabolism of lipids (including fatty acid metabolism and mitochondrial fatty acid β-oxidation), fatty acyl-CoA biosynthesis, innate immune system (including neutrophil degranulation), metabolism of vitamins and cofactors, and integration of energy metabolism (including ChREBP activates metabolic gene expression). Lipid profile analysis showed increased palmitic and palmitoleic acid content. The analysis of the data indicated that ovariectomy favored lipogenesis whereas it impaired fatty acid oxidation and induced a proinflammatory state in the visceral adipose tissue. These effects are consistent with the findings of high adiposity, hyperleptinemia, and impaired insulin sensitivity. The observed alterations were partially attenuated by estradiol replacement. The data point to a role of disrupted lipid metabolism in adipose tissue in the genesis of obesity after menopause.
Collapse
Affiliation(s)
- Valter T Boldarine
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Amanda P Pedroso
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Caroline Brandão-Teles
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Edson G LoTurco
- Divisão de Urologia e Reprodução Humana, Departamento de Cirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Cláudia M O Nascimento
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lila M Oyama
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Allain A Bueno
- Department of Biological Sciences, College of Health, Life and Environmental Sciences, University of Worcester, Worcester, United Kingdom
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION) Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, Brazil
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil
| | - Eliane B Ribeiro
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
44
|
The Role of Dysfunctional Adipose Tissue in Pancreatic Cancer: A Molecular Perspective. Cancers (Basel) 2020; 12:cancers12071849. [PMID: 32659999 PMCID: PMC7408631 DOI: 10.3390/cancers12071849] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer (PC) is a lethal malignancy with rising incidence and limited therapeutic options. Obesity is a well-established risk factor for PC development. Moreover, it negatively affects outcome in PC patients. Excessive fat accumulation in obese, over- and normal-weight individuals induces metabolic and inflammatory changes of adipose tissue microenvironment leading to a dysfunctional adipose “organ”. This may drive the association between abnormal fat accumulation and pancreatic cancer. In this review, we describe several molecular mechanisms that underpin this association at both local and systemic levels. We focus on the role of adipose tissue-derived circulating factors including adipokines, hormones and pro-inflammatory cytokines, as well as on the impact of the local adipose tissue in promoting PC. A discussion on potential therapeutic interventions, interfering with pro-tumorigenic effects of dysfunctional adipose tissue in PC, is included. Considering the raise of global obesity, research efforts to uncover the molecular basis of the relationship between pancreatic cancer and adipose tissue dysfunction may provide novel insights for the prevention of this deadly disease. In addition, these efforts may uncover novel targets for personalized interventional strategies aimed at improving the currently unsatisfactory PC therapeutic options.
Collapse
|
45
|
Gao Z, Ding R, Zhai X, Wang Y, Chen Y, Yang CX, Du ZQ. Common Gene Modules Identified for Chicken Adiposity by Network Construction and Comparison. Front Genet 2020; 11:537. [PMID: 32547600 PMCID: PMC7272656 DOI: 10.3389/fgene.2020.00537] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 05/04/2020] [Indexed: 12/12/2022] Open
Abstract
Excessive fat deposition can cause chicken health problem, and affect production efficiency by causing great economic losses to the industry. However, the molecular underpinnings of the complex adiposity trait remain elusive. In the current study, we constructed and compared the gene co-expression networks on four transcriptome profiling datasets, from two chicken lines under divergent selection for abdominal fat contents, in an attempt to dissect network compositions underlying adipose tissue growth and development. After functional enrichment analysis, nine network modules important to adipogenesis were discovered to be involved in lipid metabolism, PPAR and insulin signaling pathways, and contained hub genes related to adipogenesis, cell cycle, inflammation, and protein synthesis. Moreover, after additional functional annotation and network module comparisons, common sub-modules of similar functionality for chicken fat deposition were identified for different chicken lines, apart from modules specific to each chicken line. We further validated the lysosome pathway, and found TFEB and its downstream target genes showed similar expression patterns along with chicken preadipocyte differentiation. Our findings could provide novel insights into the genetic basis of complex adiposity traits, as well as human obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Zhuoran Gao
- College of Animal Science, Yangtze University, Jingzhou, China.,College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Ran Ding
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Xiangyun Zhai
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Yuhao Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Yaofeng Chen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Cai-Xia Yang
- College of Animal Science, Yangtze University, Jingzhou, China.,College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Zhi-Qiang Du
- College of Animal Science, Yangtze University, Jingzhou, China.,College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| |
Collapse
|
46
|
Hedbacker K, Lu YH, Dallner O, Li Z, Fayzikhodjaeva G, Birsoy K, Han C, Yang C, Friedman JM. Limitation of adipose tissue by the number of embryonic progenitor cells. eLife 2020; 9:e53074. [PMID: 32452759 PMCID: PMC7253174 DOI: 10.7554/elife.53074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 04/12/2020] [Indexed: 12/14/2022] Open
Abstract
Adipogenesis in adulthood replaces fat cells that turn over and can contribute to the development of obesity. However, the proliferative potential of adipocyte progenitors in vivo is unknown (Faust et al., 1976; Faust et al., 1977; Hirsch and Han, 1969; Johnson and Hirsch, 1972). We addressed this by injecting labeled wild-type embryonic stem cells into blastocysts derived from lipodystrophic A-ZIP transgenic mice, which have a genetic block in adipogenesis. In the resulting chimeric animals, wild-type ES cells are the only source of mature adipocytes. We found that when chimeric animals were fed a high-fat-diet, animals with low levels of chimerism showed a significantly lower adipose tissue mass than animals with high levels of chimerism. The difference in adipose tissue mass was attributed to variability in the amount of subcutaneous adipose tissue as the amount of visceral fat was independent of the level of chimerism. Our findings thus suggest that proliferative potential of adipocyte precursors is limited and can restrain the development of obesity.
Collapse
Affiliation(s)
- Kristina Hedbacker
- Laboratory of Molecular Genetics, The Rockefeller UniversityNew YorkUnited States
- Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
| | - Yi-Hsueh Lu
- Laboratory of Molecular Genetics, The Rockefeller UniversityNew YorkUnited States
| | - Olof Dallner
- Laboratory of Molecular Genetics, The Rockefeller UniversityNew YorkUnited States
| | - Zhiying Li
- Laboratory of Molecular Genetics, The Rockefeller UniversityNew YorkUnited States
| | - Gulya Fayzikhodjaeva
- Laboratory of Molecular Genetics, The Rockefeller UniversityNew YorkUnited States
- Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
| | - Kıvanç Birsoy
- Laboratory of Molecular Genetics, The Rockefeller UniversityNew YorkUnited States
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller UniversityNew YorkUnited States
| | - Chiayun Han
- Gene Targeting Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Chingwen Yang
- Gene Targeting Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, The Rockefeller UniversityNew YorkUnited States
- Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
47
|
The biology of lipid droplet-bound mitochondria. Semin Cell Dev Biol 2020; 108:55-64. [PMID: 32446655 DOI: 10.1016/j.semcdb.2020.04.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/17/2020] [Accepted: 04/18/2020] [Indexed: 12/12/2022]
Abstract
Proper regulation of cellular lipid storage and oxidation is indispensable for the maintenance of cellular energy homeostasis and health. Mitochondrial function has been shown to be a main determinant of functional lipid storage and oxidation, which is of particular interest for the adipose tissue, as it is the main site of triacylglyceride storage in lipid droplets (LDs). Recent studies have identified a subpopulation of mitochondria attached to LDs, peridroplet mitochondria (PDM) that can be separated from cytoplasmic mitochondria (CM) by centrifugation. PDM have distinct bioenergetics, proteome, cristae organization and dynamics that support LD build-up, however their role in adipose tissue biology remains largely unexplored. Therefore, understanding the molecular basis of LD homeostasis and their relationship to mitochondrial function and attachment in adipocytes is of major importance.
Collapse
|
48
|
Czech MP. Mechanisms of insulin resistance related to white, beige, and brown adipocytes. Mol Metab 2020; 34:27-42. [PMID: 32180558 PMCID: PMC6997501 DOI: 10.1016/j.molmet.2019.12.014] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/21/2019] [Accepted: 12/23/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The diminished glucose lowering effect of insulin in obesity, called "insulin resistance," is associated with glucose intolerance, type 2 diabetes, and other serious maladies. Many publications on this topic have suggested numerous hypotheses on the molecular and cellular disruptions that contribute to the syndrome. However, significant uncertainty remains on the mechanisms of its initiation and long-term maintenance. SCOPE OF REVIEW To simplify insulin resistance analysis, this review focuses on the unifying concept that adipose tissue is a central regulator of systemic glucose homeostasis by controlling liver and skeletal muscle metabolism. Key aspects of adipose function related to insulin resistance reviewed are: 1) the modes by which specific adipose tissues control hepatic glucose output and systemic glucose disposal, 2) recently acquired understanding of the underlying mechanisms of these modes of regulation, and 3) the steps in these pathways adversely affected by obesity that cause insulin resistance. MAJOR CONCLUSIONS Adipocyte heterogeneity is required to mediate the multiple pathways that control systemic glucose tolerance. White adipocytes specialize in sequestering triglycerides away from the liver, muscle, and other tissues to limit toxicity. In contrast, brown/beige adipocytes are very active in directly taking up glucose in response to β adrenergic signaling and insulin and enhancing energy expenditure. Nonetheless, white, beige, and brown adipocytes all share the common feature of secreting factors and possibly exosomes that act on distant tissues to control glucose homeostasis. Obesity exerts deleterious effects on each of these adipocyte functions to cause insulin resistance.
Collapse
Affiliation(s)
- Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
49
|
Low steady-state oxidative stress inhibits adipogenesis by altering mitochondrial dynamics and decreasing cellular respiration. Redox Biol 2020; 32:101507. [PMID: 32208164 PMCID: PMC7097524 DOI: 10.1016/j.redox.2020.101507] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/04/2020] [Accepted: 03/13/2020] [Indexed: 12/11/2022] Open
Abstract
Adipogenesis is a fundamental process of white adipose tissue function, supporting lipid storage and release, while avoiding its spillover and ectopic accumulation in tissues and organs. During aging adipogenesis is impaired and among other factors, oxidative stress contributes to this process. Adipogenesis requires functional and dynamic mitochondria; however, this organelle itself becomes dysfunctional during aging and accounts for most of reactive oxygen species (ROS) production. Here, we evaluated whether oxidative stress impairs adipogenesis through functional impairment of mitodynamics by utilizing hyperoxia as a continuous source of oxidative stress while maintaining cellular viability. This negatively impacted mitochondrial function, including respiration and dynamics and ultimately blocked adipogenesis. Interestingly, this state was reversible by using the antidiabetic drug, Rosiglitazone, which reduced oxidative stress, restored mitochondrial dynamics and respiration and augmented adipogenesis. Moreover, in vitro results were in agreement with in vivo models of oxidative stress and aging, in which mice depleted of the superoxide dismutase enzyme 1 (SOD1) and old wild-type C57BL/6JRj mice demonstrated the same trend of adipogenic potential. Importantly, in humans the results follow the same pattern, showing a downregulation of adipogenic markers during aging. Since the levels of oxidative stress and peripheral insulin resistance increase with age, while adipogenesis decreases during aging, our model helps to understand a possible way to overcome physiologically low, steady stress conditions and restore adipogenesis, avoiding accumulation of deleterious hypertrophic adipocytes in favor of beneficial hyperplasia.
Collapse
|
50
|
Wang G, Guo G, Tian X, Hu S, Du K, Zhang Q, Mao J, Jia X, Chen S, Wang J, Lai S. Screening and identification of MicroRNAs expressed in perirenal adipose tissue during rabbit growth. Lipids Health Dis 2020; 19:35. [PMID: 32145738 PMCID: PMC7060515 DOI: 10.1186/s12944-020-01219-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 03/03/2020] [Indexed: 01/26/2023] Open
Abstract
Background MicroRNAs (miRNAs) regulate adipose tissue development, which are closely related to subcutaneous and intramuscular fat deposition and adipocyte differentiation. As an important economic and agricultural animal, rabbits have low adipose tissue deposition and are an ideal model to study adipose regulation. However, the miRNAs related to fat deposition during the growth and development of rabbits are poorly defined. Methods In this study, miRNA-sequencing and bioinformatics analyses were used to profile the miRNAs in rabbit perirenal adipose tissue at 35, 85 and 120 days post-birth. Differentially expressed (DE) miRNAs between different stages were identified by DEseq in R. Target genes of DE miRNAs were predicted by TargetScan and miRanda. To explore the functions of identified miRNAs, Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed. Results Approximately 1.6 GB of data was obtained by miRNA-seq. A total of 987 miRNAs (780 known and 207 newly predicted) and 174 DE miRNAs were identified. The miRNAs ranged from 18 nt to 26 nt. GO enrichment and KEGG pathway analyses revealed that the target genes of the DE miRNAs were mainly involved in zinc ion binding, regulation of cell growth, MAPK signaling pathway, and other adipose hypertrophy-related pathways. Six DE miRNAs were randomly selected, and their expression profiles were validated by q-PCR. Conclusions This is the first report of the miRNA profiles of adipose tissue during different growth stages of rabbits. Our data provide a theoretical reference for subsequent studies on rabbit genetics, breeding and the regulatory mechanisms of adipose development.
Collapse
Affiliation(s)
- Guoze Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211# Huimin Road, Wenjiang, 611130, Sichuan, China.,Guizhou Medical University, Guiyang, 550025, China
| | - Guo Guo
- Guizhou Medical University, Guiyang, 550025, China
| | - Xueting Tian
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211# Huimin Road, Wenjiang, 611130, Sichuan, China
| | - Kun Du
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211# Huimin Road, Wenjiang, 611130, Sichuan, China
| | | | - Jingxin Mao
- Southwest University, Chongqing, 400715, China
| | - Xianbo Jia
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211# Huimin Road, Wenjiang, 611130, Sichuan, China
| | - Shiyi Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211# Huimin Road, Wenjiang, 611130, Sichuan, China
| | - Jie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211# Huimin Road, Wenjiang, 611130, Sichuan, China
| | - Songjia Lai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211# Huimin Road, Wenjiang, 611130, Sichuan, China.
| |
Collapse
|