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; 21:272-288. [PMID: 39757324 DOI: 10.1038/s41574-024-01071-y] [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: 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
|
Ahn C, Divoux A, Zhou M, Seldin MM, Sparks LM, Whytock KL. Optimized RNA sequencing deconvolution illustrates the impact of obesity and weight loss on cell composition of human adipose tissue. Obesity (Silver Spring) 2025; 33:936-948. [PMID: 40176378 DOI: 10.1002/oby.24264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 04/04/2025]
Abstract
OBJECTIVE Cellular heterogeneity of human adipose tissue is linked to the pathophysiology of obesity and may impact the response to energy restriction and changes in fat mass. Herein, we provide an optimized pipeline to estimate cellular composition in human abdominal subcutaneous adipose tissue (ASAT) bulk RNA sequencing (RNA-seq) datasets using a single-nuclei RNA-seq signature matrix. METHODS A deconvolution pipeline for ASAT was optimized by benchmarking publicly available algorithms using a signature matrix derived from ASAT single-nuclei RNA-seq data from 20 adults and then applied to estimate ASAT cell-type proportions in publicly available obesity and weight loss studies. RESULTS Individuals with obesity had greater proportions of macrophages and lower proportions of adipocyte subpopulations and vascular cells compared with lean individuals. Two months of diet-induced weight loss increased the estimated proportions of macrophages; however, 2 years of diet-induced weight loss reduced the estimated proportions of macrophages, thereby suggesting a biphasic nature of cellular remodeling of ASAT during weight loss. CONCLUSIONS Our optimized high-throughput pipeline facilitates the assessment of composition changes of highly characterized cell types in large numbers of ASAT samples using low-cost bulk RNA-seq. Our data reveal novel changes in cellular heterogeneity and its association with cardiometabolic health in humans with obesity and following weight loss.
Collapse
Affiliation(s)
- Cheehoon Ahn
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| | - Adeline Divoux
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| | - Mingqi Zhou
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, California, USA
| | - Marcus M Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, California, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| | - Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| |
Collapse
|
3
|
Asgari S, Masrouri S, Khalili D, Lotfaliany M, Hadaegh F. Weight Gain, Weight Loss, and Type 2 Diabetes Risk: Evidence From the Atherosclerosis Risk in Communities (ARIC) Study. Endocrinol Diabetes Metab 2025; 8:e70040. [PMID: 40198876 PMCID: PMC11978231 DOI: 10.1002/edm2.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 02/02/2025] [Accepted: 02/22/2025] [Indexed: 04/10/2025] Open
Abstract
INTRODUCTION While type 2 diabetes (T2DM) has become a major health issue in the North American and Caribbean region, the effects of weight change on incident T2DM, conditional on either initial or attained weight, are poorly addressed. Therefore, we aimed to assess the impact of 3-year weight change on incident T2DM over 6 years among US individuals. METHODS A total of 8377 participants aged 45-64 years (4601 women), free of T2DM or cancer at baseline from the Atherosclerosis Risk in Communities (ARIC) study were included. Weight measurements were taken at baseline (visit 1, 1987-89) and approximately 3 years later (visit 2, 1990-92). Participants were categorised based on their weight change ratio into ≥ 5% weight loss, stable (±5%), and ≥ 5% weight gain. Cox proportional hazards models, adjusting for known diabetes risk factors, were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) of incident T2DM, with stable weight (±5%) as the reference category. RESULTS During a median follow-up period of 6 years, participants were classified into three categories: 361 persons remained stable (±5%), 47 with ≥ 5% loss, and 135 with ≥ 5% gain. In multivariable analysis, after adjustment with initial weight, ≥ 5% weight gain and loss were significantly associated with higher [HR (95% CI): 1.68 (1.36-2.06), p-value < 0.0001] and lower [0.73 (0.53-1.00), p-value = 0.05] risks of incident T2DM, respectively. When adjusted for attained weight, weight gain ≥ 5% remained a significant risk factor for T2DM [1.51 (1.21-1.88)]; however, weight loss ≥ 5% lost statistical significance [0.84 (0.60-1.17), p-value = 0.31]. CONCLUSIONS We found a robust association between weight gain and incident T2DM; however, the beneficial impact of weight loss was significantly attenuated after considering the attained weight.
Collapse
Affiliation(s)
- Samaneh Asgari
- Prevention of Metabolic Disorders Research Center, Research Institute for Metabolic and Obesity Disorders, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Soroush Masrouri
- Prevention of Metabolic Disorders Research Center, Research Institute for Metabolic and Obesity Disorders, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Davood Khalili
- Prevention of Metabolic Disorders Research Center, Research Institute for Metabolic and Obesity Disorders, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Mojtaba Lotfaliany
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon HealthDeakin UniversityGeelongAustralia
| | - Farzad Hadaegh
- Prevention of Metabolic Disorders Research Center, Research Institute for Metabolic and Obesity Disorders, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
4
|
Kurihara Y, Shimizu A, Ozuru R, Yoshimura M, Chou B, Itoh R, Ishii K, Hirota Y, Takagi S, Fujita M, Inoue M, Hiromatsu K. Mycobacterium abscessus resides within lipid droplets and acquires a dormancy-like phenotype in adipocytes. Biochem Biophys Res Commun 2025; 758:151645. [PMID: 40120350 DOI: 10.1016/j.bbrc.2025.151645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Mycobacterium abscessus (M. abscessus) is an emerging, rapidly growing mycobacterium that causes chronic lung infection, particularly in patients with cystic fibrosis, as well as skin and soft tissue infections. Adipose tissue is recognized as an important niche that supports M. tuberculosis persistence. However, the dormancy, latency, and persistence mechanisms of M. abscessus in the host remain poorly understood. This study investigated how adipose tissue serves as a niche for M. abscessus using both a human adipose tissue ex vivo infection model and a murine adipose tissue in vivo infection model. M. abscessus infected not only the cytosol of adipocytes but also entered host lipid droplets, where it formed intracytoplasmic lipid inclusions in the bacterial cell. To our knowledge, this unique localization has never been reported for M. abscessus or any other mycobacterium. Within host lipid droplets, M. abscessus lost acid-fastness and gained Nile Red positivity. These results suggest that M. abscessus acquires a dormancy-like phenotype within host lipid droplets of adipocytes, potentially contributing to its persistence, virulence, and resistance to treatment. These findings provide valuable insights into mycobacterial persistence mechanisms and could offer a promising foundation for developing novel therapeutic approaches.
Collapse
Affiliation(s)
- Yusuke Kurihara
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan; Department of Infectious Medicine Division of Eukaryotic Microbiology, Faculty of Medicine, Kurume University, Fukuoka, 830-0011, Japan.
| | - Akinori Shimizu
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Ryo Ozuru
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Michinobu Yoshimura
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Bin Chou
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Ryota Itoh
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Kazunari Ishii
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Yuko Hirota
- Department of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Satoshi Takagi
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Masaki Fujita
- Department of Respiratory Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Masahiro Inoue
- Department of Infectious Medicine Division of Eukaryotic Microbiology, Faculty of Medicine, Kurume University, Fukuoka, 830-0011, Japan
| | - Kenji Hiromatsu
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| |
Collapse
|
5
|
Wing A, Jeffery E, Church CD, Goodell J, Saavedra-Peña RDM, Saha M, Holtrup B, Voisin M, Alavi NS, Floody M, Wang Z, Zapadka TE, Garabedian MJ, Varshney R, Rudolph MC, Rodeheffer MS. Dietary oleic acid drives obesogenic adipogenesis via modulation of LXRα signaling. Cell Rep 2025; 44:115527. [PMID: 40208790 DOI: 10.1016/j.celrep.2025.115527] [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: 08/06/2024] [Revised: 02/17/2025] [Accepted: 03/15/2025] [Indexed: 04/12/2025] Open
Abstract
Dietary fat composition has changed substantially during the obesity epidemic. As adipocyte hyperplasia is a major mechanism of adipose expansion, we aim to ascertain how dietary fats affect adipogenesis during obesity. We employ an unbiased dietary screen to identify oleic acid (OA) as the only dietary fatty acid that induces obesogenic hyperplasia at physiologic levels and show that plasma monounsaturated fatty acids (MUFAs), which are mostly OA, are associated with human obesity. OA stimulates adipogenesis in mouse and human adipocyte precursor cells (APCs) by increasing AKT2 signaling, a hallmark of obesogenic hyperplasia, and reducing LXR activity. High OA consumption decreases LXRα Ser196 phosphorylation in APCs, while blocking LXRα phosphorylation results in APC hyperproliferation. As OA is increasingly being incorporated into dietary fats due to purported health benefits, our finding that OA is a unique physiologic regulator of adipose biology underscores the importance of understanding how high OA consumption affects metabolic health.
Collapse
Affiliation(s)
- Allison Wing
- Department of Molecular, Cell, and Developmental Biology, Yale University, 219 Prospect St., New Haven, CT 06520, USA
| | - Elise Jeffery
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06520, USA
| | - Christopher D Church
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., New Haven, CT 06520, USA
| | - Jennifer Goodell
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., New Haven, CT 06520, USA
| | - Rocío Del M Saavedra-Peña
- Department of Molecular, Cell, and Developmental Biology, Yale University, 219 Prospect St., New Haven, CT 06520, USA
| | - Moumita Saha
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., New Haven, CT 06520, USA
| | - Brandon Holtrup
- Department of Molecular, Cell, and Developmental Biology, Yale University, 219 Prospect St., New Haven, CT 06520, USA
| | - Maud Voisin
- Department of Microbiology, NYU School of Medicine, New York, NY 10016, USA
| | - N Sima Alavi
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., New Haven, CT 06520, USA
| | - Mariana Floody
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., New Haven, CT 06520, USA
| | - Zenan Wang
- Department of Molecular, Cell, and Developmental Biology, Yale University, 219 Prospect St., New Haven, CT 06520, USA
| | - Thomas E Zapadka
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Michael J Garabedian
- Department of Microbiology, NYU School of Medicine, New York, NY 10016, USA; Department of Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Rohan Varshney
- Department of Biochemistry and Physiology and Harold Hamm Diabetes Center, Oklahoma University Health Sciences, Oklahoma City, OK 73104, USA
| | - Michael C Rudolph
- Department of Biochemistry and Physiology and Harold Hamm Diabetes Center, Oklahoma University Health Sciences, Oklahoma City, OK 73104, USA.
| | - Matthew S Rodeheffer
- Department of Molecular, Cell, and Developmental Biology, Yale University, 219 Prospect St., New Haven, CT 06520, USA; Department of Cell Biology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06520, USA; Yale Stem Cell Center, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06520, USA; Yale Center of Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
6
|
Emílio-Silva MT, Rodrigues VP, Fioravanti MM, Ruiz-Malagon AJ, Naia Fioretto M, Raimundo PR, Ohara R, Assunção R, Bueno G, Dario FL, Justulin LA, Rodríguez-Nogales A, da Rocha LRM, Gálvez J, Hiruma-Lima CA. Citral protects against metabolic endotoxemia, and systemic disorders caused by high-fat diet-induced obesity via intestinal modulation. Front Pharmacol 2025; 16:1567217. [PMID: 40260376 PMCID: PMC12009827 DOI: 10.3389/fphar.2025.1567217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Background Obesity is a growing global epidemic associated with changes in the gut microenvironment and metabolic endotoxemia, which can exacerbate metabolic and inflammatory processes. Citral (CT), a monoterpene present in essential oils, has been investigated for its anti-inflammatory, antioxidant, and immunomodulatory properties. However, its role in modulating the gut axis during metabolic and inflammatory alterations in obesity remains unknown. In this study, we investigated the effects of CT on intestinal and metabolic impairment induced by lipopolysaccharide (LPS) and high-fat diet (HFD) in in vitro and in vivo models. Methods Male C57BL/6J mice were fed a standard diet and HFD for 17 weeks, with daily oral administration of CT treatment (25, 100, or 300 mg/kg) or vehicle. Morphological and histological parameters, lipid profiles, adipose index, cytokine levels, and colonic gene expression were determined. In vitro, murine rectal carcinoma (CMT-93) cells were stimulated with LPS (10 μg/mL) to assess tight junction and inflammatory protein expression. Results CT treatment showed anti-obesity activity against HFD-induced body mass gain in mice, which was attributed to a significant reduction in body fat, glycemia, and cholesterol levels. Systemic inflammation during obesity also decreased after CT treatment, with a significant reduction in serum levels of endotoxin, interleukin-1β, and tumor necrosis factor-α. Additionally, CT stimulation reduced inducible nitric oxide synthase expression and maintained ZO-1 levels in LPS-stimulated CMT-93 cells. Conclusion CT has anti-obesogenic, anti-inflammatory, and anti-hyperlipidemic properties mediated by its protective effects on the intestinal epithelium in obesity. Thus, our results highlight the promising preclinical results of CT treatment as a protective agent against the detrimental effects of HFD and LPS in mice.
Collapse
Affiliation(s)
- Maycon Tavares Emílio-Silva
- Department of Structural and Functional Biology, Physiology Sector, Institute of Bioscience, São Paulo State University (UNESP), Botucatu, Brazil
| | - Vinicius Peixoto Rodrigues
- Department of Structural and Functional Biology, Physiology Sector, Institute of Bioscience, São Paulo State University (UNESP), Botucatu, Brazil
| | - Mariana Moraes Fioravanti
- Department of Structural and Functional Biology, Physiology Sector, Institute of Bioscience, São Paulo State University (UNESP), Botucatu, Brazil
| | - Antonio Jesús Ruiz-Malagon
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Matheus Naia Fioretto
- Department of Structural and Functional Biology, Morphology Sector, Institute of Bioscience, São Paulo State University, (UNESP), Botucatu, Brazil
| | - Priscila Romano Raimundo
- Department of Structural and Functional Biology, Physiology Sector, Institute of Bioscience, São Paulo State University (UNESP), Botucatu, Brazil
| | - Rie Ohara
- Department of Structural and Functional Biology, Physiology Sector, Institute of Bioscience, São Paulo State University (UNESP), Botucatu, Brazil
| | - Renata Assunção
- Department of Structural and Functional Biology, Physiology Sector, Institute of Bioscience, São Paulo State University (UNESP), Botucatu, Brazil
| | - Gabriela Bueno
- Department of Structural and Functional Biology, Physiology Sector, Institute of Bioscience, São Paulo State University (UNESP), Botucatu, Brazil
| | - Felipe Lima Dario
- Department of Structural and Functional Biology, Physiology Sector, Institute of Bioscience, São Paulo State University (UNESP), Botucatu, Brazil
| | - Luis Antonio Justulin
- Department of Structural and Functional Biology, Morphology Sector, Institute of Bioscience, São Paulo State University, (UNESP), Botucatu, Brazil
| | - Alba Rodríguez-Nogales
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Lucia Regina Machado da Rocha
- Department of Structural and Functional Biology, Physiology Sector, Institute of Bioscience, São Paulo State University (UNESP), Botucatu, Brazil
| | - Júlio Gálvez
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- CIBER de Enfermedades Hepáticas y Digestivas (CIBER-EHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Clélia Akiko Hiruma-Lima
- Department of Structural and Functional Biology, Physiology Sector, Institute of Bioscience, São Paulo State University (UNESP), Botucatu, Brazil
| |
Collapse
|
7
|
Liu B, Yao Z, Song L, Sun C, Shen C, Cheng F, Cheng Z, Zhang R, Liu R. Vitexin alleviates lipid metabolism disorders and hepatic injury in obese mice through the PI3K/AKT/mTOR/SREBP-1c pathway. Eur J Med Chem 2025; 287:117379. [PMID: 39947052 DOI: 10.1016/j.ejmech.2025.117379] [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: 11/21/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/24/2025]
Abstract
Obesity is recognized as a metabolic disorder, and its treatment and management pose ongoing challenges worldwide. Hawthorn, a traditional Chinese herb used to alleviate digestive issues and reduce blood lipid levels, has unclear mechanisms of action regarding its active components in the treatment of obesity. This study investigated the anti-obesity effects of vitexin, a major flavonoid compound found in hawthorn, in high-fat diet (HFD)-induced C57BL/6 mice. The results demonstrated that vitexin significantly reduced body weight, liver weight, blood lipid levels, and inflammatory markers in obese mice, while also inhibiting hepatic lipid accumulation. Mechanistic studies revealed that vitexin likely suppresses adipogenesis by modulating the PI3K-AKT signaling pathway, as evidenced by reduced expression of PI3K, phosphorylated AKT, phosphorylated mTOR, and SREBP-1c in the livers of vitexin-treated obese mice. Additionally, vitexin inhibited NFκB expression by regulating IκBα phosphorylation, thereby alleviating obesity-induced liver injury. These findings suggest that vitexin may be the primary active component in hawthorn responsible for reducing blood lipid levels, highlighting its potential in the treatment of obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
- Bo Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ziqing Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lin Song
- Department of Pharmacy, Children' S Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Chen Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Changhong Shen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zefang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ruoqi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Rong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
8
|
Zhao Y, Yue R. White adipose tissue in type 2 diabetes and the effect of antidiabetic drugs. Diabetol Metab Syndr 2025; 17:116. [PMID: 40186308 PMCID: PMC11969724 DOI: 10.1186/s13098-025-01678-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 03/19/2025] [Indexed: 04/07/2025] Open
Abstract
White adipose tissue (WAT) is highly flexible and was previously considered a passive location for energy storage. Its endocrine function has been established for several years, earning it the title of an "endocrine organ" due to its ability to secrete many adipokines that regulate metabolism. WAT is one of the core tissues that influence insulin sensitivity. Its dysfunction enhances insulin resistance and type 2 diabetes (T2D) progression. However, T2D may cause WAT dysfunction, including changes in distribution, metabolism, adipocyte hypertrophy, inflammation, aging, and adipokines and free fatty acid levels, which may exacerbate insulin resistance. This review used PubMed to search WAT dysfunction in T2D and the effects of these changes on insulin resistance. Additionally, we described and discussed the effects of antidiabetic drugs, including insulin therapy, sulfonylureas, metformin, glucose-like peptide-1 receptor agonists, thiazolidinediones, and sodium-dependent glucose transporters-2 inhibitors, on WAT parameters under T2D conditions.
Collapse
Affiliation(s)
- Yixuan Zhao
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu, University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan Province, 610072, P. R. China
| | - Rensong Yue
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu, University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan Province, 610072, P. R. China.
| |
Collapse
|
9
|
Domagalski M, Olszańska J, Pietraszek‐Gremplewicz K, Nowak D. The role of adipogenic niche resident cells in colorectal cancer progression in relation to obesity. Obes Rev 2025; 26:e13873. [PMID: 39763022 PMCID: PMC11884973 DOI: 10.1111/obr.13873] [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: 10/17/2023] [Revised: 10/03/2024] [Accepted: 11/05/2024] [Indexed: 03/08/2025]
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide and has one of the highest mortality rates. Considering its nonlinear etiology, many risk factors are associated with CRC formation and development, with obesity at the forefront. Obesity is regarded as one of the key environmental risk determinants for the pathogenesis of CRC. Excessive food intake and a sedentary lifestyle, together with genetic predispositions, lead to the overgrowth of adipose tissue along with a disruption in the number and function of its building cells. Adipose tissue-resident cells may constitute part of the CRC microenvironment. Alterations in their physiology and secretory profiles observed in obesity may further contribute to CRC progression, and despite similar localization, their contributions are not equivalent. They can interact with CRC cells, either directly or indirectly, influencing various processes that contribute to tumorigenesis. The main aim of this review is to provide insights into the diversity of adipose tissue resident cells, namely, adipocytes, adipose stromal cells, and immunological cells, regarding the role of particular cell types in co-forming the CRC microenvironment. The scope of this study was also devoted to the abnormalities in adipose tissue physiology observed in obesity states and their impact on CRC development.
Collapse
Affiliation(s)
- Mikołaj Domagalski
- Department of Cell Pathology, Faculty of BiotechnologyUniversity of WroclawWroclawPoland
| | - Joanna Olszańska
- Department of Cell Pathology, Faculty of BiotechnologyUniversity of WroclawWroclawPoland
| | | | - Dorota Nowak
- Department of Cell Pathology, Faculty of BiotechnologyUniversity of WroclawWroclawPoland
| |
Collapse
|
10
|
Neuwald NV, Pearce AL, Cunningham PM, Setzenfand MN, Koczwara L, Rolls BJ, Keller KL. Food switching at a meal is positively associated with change in adiposity among children at high-familial risk for obesity. Appetite 2025; 208:107915. [PMID: 40010570 PMCID: PMC11948248 DOI: 10.1016/j.appet.2025.107915] [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: 10/25/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
Switching between different foods while eating has been positively associated with weight status and intake in children. Evidence suggests that switching behavior is consistent within children across meals, however, it is unclear how switching relates to changes in adiposity over time. In a 1-year longitudinal study, we assessed whether food switching predicted changes in fat mass index (FMI: fat mass kg/height m2) in 7-8-year-old children and tested if familial risk of obesity moderated this relationship. At baseline, seventy-four children without obesity (7.8 ± 0.6 y; 37F) consumed four ad libitum meals of varying portion sizes, each consisting of chicken nuggets, macaroni and cheese, grapes, and broccoli. For each child, the average number of food switches was calculated from video recordings across the four meals. To assess change in adiposity over time, children completed a dual energy x-ray absorptiometry scan for assessment of FMI at baseline and follow-up (≥1 year later). Familial risk of obesity was determined by maternal BMI (high-risk: ≥30 kg/m2, n = 32 vs. low-risk: <25 kg/m2, n = 42). Food switching at baseline was positively associated with changes in FMI over 1 year (p = 0.03). In addition to the 37% of variance in FMI change explained by known factors influencing adiposity, food switching accounted for an additional 4% of the variance (p = 0.03). Further, there was an interaction between familial risk status and food switching (p = 0.04) such that the relationship between switching and FMI change was only significant in high-risk children. Overall, children's food switching behavior assessed at laboratory meals predicted change in adiposity over 1 year. Food switching could be a behavioral marker for, and contribute to, pediatric obesity risk particularly in children with a familial predisposition.
Collapse
Affiliation(s)
- Nicholas V Neuwald
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Alaina L Pearce
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Paige M Cunningham
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA; Department of Food Science, The Pennsylvania State University, University Park, PA, USA
| | - Marissa N Setzenfand
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Lauren Koczwara
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Barbara J Rolls
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Kathleen L Keller
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA; Department of Food Science, The Pennsylvania State University, University Park, PA, USA; Social Science Research Institute, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
11
|
Arner P, Sørensen TI, Andersson DP. Adipose cellularity as a measurement of long-term changes in body weight: a Swedish cohort study spanning 1988-2016. EClinicalMedicine 2025; 82:103165. [PMID: 40235948 PMCID: PMC11997358 DOI: 10.1016/j.eclinm.2025.103165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 04/17/2025] Open
Abstract
Background Adipocyte size and number (cellularity) determine the adipose mass and may relate to long-term body weight changes. Methods We investigated 1014 healthy participants at Karolinska Institutet in Sweden 1988-2016 for body weight and size/number of subcutaneous adipocytes, and 273 for visceral adipocyte size. We measured body weight on 281 subjects about 16 years later. We analysed the association of baseline adipocyte size and number with body weight changes by linear regression including relevant co-factors, and the associations of cellularity (low or high number of either large or small adipocytes) regarding body weight changes by analysis of variance. Findings Subcutaneous adipocyte size and number and visceral adipocyte size showed strong relationships with body weight changes irrespective of its mode of expression (adjusted r2 ≥0·15). The relationships were significant (p ≤ 0·027) independent of co-factors (age, sex, initial body weight or height, body fat, obesity, nicotine use, physical activity, and observation time). Interventions (lifestyle change or bariatric surgery) did not influence the associations (p = 0·86). A low or high number of large adipocytes associated with body weight loss, whereas a low or high number of small cells associated with weight stability or weight gain. Interpretation Adipose cellularity is associated with long-term changes in body weight, following interventions to decrease body weight. Patients with a high number of large fat cells experienced the most pronounced weight reduction. Funding The Stockholm County Council (963296, 994175, 986118), the Center for Innovative Medicine at Karolinska Institutet (986109) and the Swedish Society of Medicine (1001156). None of the funding sources had any involvement in the study.
Collapse
Affiliation(s)
- Peter Arner
- Department of Medicine-H7 at Karolinska Institutet, C2:94 Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| | - Thorkild I.A. Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Public Health Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen, 2200N, Denmark
- Center for Childhood Health, Islands Brygge 41, Copenhagen, 2300S, Denmark
| | - Daniel P. Andersson
- Department of Medicine-H7 at Karolinska Institutet, C2:94 Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
- Department of Endocrinology, C2:94 Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| |
Collapse
|
12
|
Cheong L, Law LSC, Tan LYL, Amal AAA, Khoo CM, Eng PC. Medical Nutrition Therapy for Women with Gestational Diabetes: Current Practice and Future Perspectives. Nutrients 2025; 17:1210. [PMID: 40218968 PMCID: PMC11990351 DOI: 10.3390/nu17071210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Gestational diabetes mellitus (GDM) is a complication that affects 20% of pregnancies worldwide. It is associated with adverse short- and long-term cardiometabolic outcomes for both mother and infant. Effective management of GDM involves lifestyle modifications, including medical nutrition therapy (MNT) and physical activity (PA), with the addition of insulin or metformin if glycaemic control remains inadequate. However, substantial gaps persist in the determination of optimal medical nutrition therapy (MNT) for women with GDM. Challenges in MNT include individual variation in glucose tolerance and changing maternal physiology and dietary requirements during pregnancy. Achieving optimal glycaemic control depends on careful macronutrient balance, particularly the distribution and quality of carbohydrate intake and sufficient protein and fat intake. Additionally, micronutrient deficiencies, such as inadequate vitamin D, calcium, and essential minerals, may exacerbate oxidative stress, inflammation, and glycaemic dysregulation, further impacting foetal growth and development. Cultural beliefs and dietary practices among pregnant women can also hinder adherence to recommended nutritional guidelines. Conditions like hyperemesis gravidarum (HG) affect ~1% to 2% of pregnant women can result in unintended energy and nutrient deficits. This special issue explores the current evidence and major barriers to optimising dietary therapy for women with GDM. It also identifies future research priorities to advance clinical practice, improve maternal and foetal outcomes, and address gaps in personalised nutrition interventions.
Collapse
Affiliation(s)
- Louisa Cheong
- Division of Endocrinology, Department of Medicine, National University Hospital, Singapore 119074, Singapore; (L.C.); (L.S.-C.L.); (L.Y.L.T.); (C.M.K.)
| | - Lawrence Siu-Chun Law
- Division of Endocrinology, Department of Medicine, National University Hospital, Singapore 119074, Singapore; (L.C.); (L.S.-C.L.); (L.Y.L.T.); (C.M.K.)
| | - Li Ying Lyeann Tan
- Division of Endocrinology, Department of Medicine, National University Hospital, Singapore 119074, Singapore; (L.C.); (L.S.-C.L.); (L.Y.L.T.); (C.M.K.)
| | - Amal Al-Amri Amal
- Department of Internal Medicine, Nizwa Hospital, Nizwa P.O. Box 1222, Oman;
| | - Chin Meng Khoo
- Division of Endocrinology, Department of Medicine, National University Hospital, Singapore 119074, Singapore; (L.C.); (L.S.-C.L.); (L.Y.L.T.); (C.M.K.)
| | - Pei Chia Eng
- Division of Endocrinology, Department of Medicine, National University Hospital, Singapore 119074, Singapore; (L.C.); (L.S.-C.L.); (L.Y.L.T.); (C.M.K.)
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| |
Collapse
|
13
|
Kamrul‐Hasan ABM, Pappachan JM, Nagendra L, Ashraf H, Dutta D, Bhattacharya S, Kapoor N. Metabolic outcomes of bariatric surgery versus lifestyle intervention in adolescents with severe obesity: A systematic review and meta‐analysis. Clin Obes 2025. [DOI: 10.1111/cob.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/25/2025] [Indexed: 03/31/2025]
Abstract
SummaryData from clinical trials evaluating the effectiveness and safety of metabolic and bariatric surgery (MBS) compared to lifestyle modifications (LSM) in children and adolescents with obesity are scarce. This systematic review and meta‐analysis (SRM) sought to fill this knowledge gap. Randomised or non‐randomised trials spanning at least one‐year involving children and adolescents with severe obesity receiving any form of MBS in the intervention group and LSM for weight loss in the control group were systematically searched through electronic databases. This SRM adhered to the guidelines outlined in the Cochrane Handbook for Systematic Reviews of Interventions and the PRISMA checklists. The primary outcome of interest was the change in body weight from the baseline. Five trials (three randomised, open‐label and two non‐randomised) with 1–2 years follow‐up durations were analysed, including 367 participants aged 10–19 years. MBS resulted in greater reductions in body weight (mean difference [MD] −25.83 kg, 95% confidence interval [CI] [−36.91, −14.75], p < .00001) and per cent body weight (MD −24.54%, 95% CI [−33.19, −15.89], p < .00001) compared to LSM. Furthermore, MBS outperformed LSM in reducing body mass index (BMI), BMI z‐score, waist circumference, glycated haemoglobin, fasting plasma glucose, insulin resistance, triglycerides, alanine aminotransferase, high sensitivity C‐reactive protein and an overall improvement of physical functioning and quality of life. The safety profile was comparable between the two groups; however, data was scarce. Larger, longer‐term trials that include multinational and multiethnic representation are essential for making solid clinical practice recommendations regarding MBS for children with obesity.
Collapse
Affiliation(s)
| | - Joseph M. Pappachan
- Faculty of Science Manchester Metropolitan University Manchester UK
- Department of Endocrinology Kasturba Medical College, Manipal Academy of Higher Education Manipal India
| | - Lakshmi Nagendra
- Department of Endocrinology JSS Medical College, JSS Academy of Higher Education and Research Mysore Karnataka India
| | - Hamid Ashraf
- Department of Endocrinology and Diabetes Rajiv Gandhi Centre of Diabetes and Endocrinology, Aligarh Muslim University Aligarh India
| | - Deep Dutta
- Department of Endocrinology CEDAR Superspeciality Healthcare New Delhi India
| | | | - Nitin Kapoor
- Department of Endocrinology, Diabetes and Metabolism Christian Medical College Vellore Tamil Nadu India
- Non‐Communicable Disease Unit, Melbourne School of Population and Global Health University of Melbourne Carlton Victoria Australia
| |
Collapse
|
14
|
Al Dow M, Secco B, Mouchiroud M, Rochette M, Gilio GR, Massicard M, Hardy M, Gélinas Y, Festuccia WT, Morissette MC, Manem VSK, Laplante M. Loss of VSTM2A promotes adipocyte hypertrophy and disrupts metabolic homeostasis. Obesity (Silver Spring) 2025; 33:522-536. [PMID: 39956640 PMCID: PMC11897849 DOI: 10.1002/oby.24224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/16/2024] [Accepted: 11/05/2024] [Indexed: 02/18/2025]
Abstract
OBJECTIVE Adipose tissue expands through hyperplasia and hypertrophy to store excess lipids, a process that is essential for the maintenance of metabolic homeostasis. The mechanisms regulating adipocyte recruitment from progenitors remain unclear. We have previously identified V-set and transmembrane domain-containing protein 2A (VSTM2A) as a factor promoting fat cell development in vitro. Whether VSTM2A impacts adipose tissue and systemic metabolism in vivo is still unknown. METHODS We generated VSTM2A knockout mice (Vstm2a-/-) using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) and fed them either a chow or high-fat diet. These mice were evaluated for body weight, adiposity, blood parameters, and glucose homeostasis. RESULTS Vstm2a-/- mice were viable and showed no body weight differences. Although adipose mass was similar, Vstm2a-/- mice had larger adipocytes, an effect linked to inflammation, ectopic lipid deposition, and impaired glucose and lipid metabolism. Transcriptomic analysis revealed that VSTM2A loss affects the expression of several genes in adipose tissue, including some related to the lysosome. Interestingly, acute lysosomal inhibition early in life is sufficient to cause adipocyte hypertrophy in adults. CONCLUSIONS VSTM2A is dispensable for adipose tissue formation, but its loss causes adipocyte hypertrophy and impairs glucose and lipid homeostasis. Our study also underscores a critical role of the lysosome in initiating adipogenesis.
Collapse
Affiliation(s)
- Manal Al Dow
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
| | - Blandine Secco
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
| | - Mathilde Mouchiroud
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
| | - Marianne Rochette
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
| | - Gustavo R. Gilio
- Institute of Biomedical Sciences, Department of Physiology and BiophysicsUniversity of São PauloSão PauloBrazil
| | - Mickael Massicard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
| | - Marilou Hardy
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
| | - Yves Gélinas
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
| | - William T. Festuccia
- Institute of Biomedical Sciences, Department of Physiology and BiophysicsUniversity of São PauloSão PauloBrazil
| | - Mathieu C. Morissette
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
- Centre de recherche sur le cancer de l'Université LavalUniversité LavalQuebecQuébec CityCanada
- Département de Médecine, Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
| | - Venkata S. K. Manem
- Centre de recherche sur le cancer de l'Université LavalUniversité LavalQuebecQuébec CityCanada
- Département de Médecine, Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
- Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
| | - Mathieu Laplante
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
- Centre de recherche sur le cancer de l'Université LavalUniversité LavalQuebecQuébec CityCanada
- Département de Médecine, Faculté de MédecineUniversité LavalQuebecQuébec CityCanada
| |
Collapse
|
15
|
Bench G. The development and evolution of biological AMS at Livermore: a perspective. Bioanalysis 2025; 17:345-354. [PMID: 39902785 PMCID: PMC11875510 DOI: 10.1080/17576180.2025.2460391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025] Open
Abstract
Biological accelerator mass spectrometry (AMS) provides ultrasensitive carbon-14 isotopic analysis enabling a deeper understanding of human health concerns by enabling quantification of pharmacokinetics and other molecular endpoints directly in humans. It enables environmentally and human relevant studies of metabolic pathways through the use of very low concentrations of labeled metabolic substrates in cells and organisms. Here, we discuss why AMS is an important tool for the biosciences, the development and evolution of biological AMS at Livermore and discuss technical refinements that will improve the efficiency of operation for the measurement of ultra-trace levels of 14C, which, long term, will enable greater ease of use and sample throughput.
Collapse
Affiliation(s)
- Graham Bench
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA
| |
Collapse
|
16
|
Scheidl TB, Wager JL, Thompson JA. Adipose Tissue Stromal Cells: Rheostats for Adipose Tissue Function and Metabolic Disease Risk. Can J Cardiol 2025:S0828-282X(25)00137-0. [PMID: 39986382 DOI: 10.1016/j.cjca.2025.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025] Open
Abstract
The transition from metabolically healthy obesity to the development of obesity-associated metabolic syndrome and cardiovascular disease is thought to be triggered by a loss in the functional integrity of adipose tissue. Although mature adipocytes are the primary functional units that carry out lipid partitioning in adipose tissue for the promotion of whole-body energy balance, they are supported by a heterogenous collection of nonadipocytes in the stroma. Research over the past couple of decades has expanded perspectives on the homeostatic and pathological roles of the nonadipocyte compartment. Adipose progenitors originate in the embryonic period and drive the developmental adipogenesis that establishes the set point of adiposity. A population of adipocyte progenitors reside in adult depots and serve an important homeostatic role as a reservoir to support adipocyte turnover. Adipocyte hypertrophy in obesity increases the rate of adipocyte death and the ability of progenitors to support this high rate of adipocyte turnover is important for the preservation of the lipid-buffering function of adipose tissue. Some evidence exists to suggest that impaired adipogenesis or a decline in progenitors capable of differentiation is a key event in the development of adipose dysfunction. The efficiency of macrophages to clear the debris and toxic lipids released from dead adipocytes lies at the fulcrum between preservation of adipose function and the progression toward chronic inflammation. Although macrophages in collaboration with other immune cells propagate the inflammation that underlies adipose dysfunction, there is now a greater appreciation for the diverse and unique roles of immune cells within adipose tissue.
Collapse
Affiliation(s)
- Taylor B Scheidl
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada. https://twitter.com/TaylorScheidl
| | - Jessica L Wager
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jennifer A Thompson
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
17
|
Jørsboe E, Kubitz P, Honecker J, Flaccus A, Mvondo D, Raggi M, Hansen T, Hauner H, Blüher M, Charles PD, Lindgren CM, Nellåker C, Claussnitzer M. Deep Learning Derived Adipocyte Size Reveals Adipocyte Hypertrophy is under Genetic Control. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.11.25322053. [PMID: 39990583 PMCID: PMC11844614 DOI: 10.1101/2025.02.11.25322053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Fat distribution and macro structure of white adipose tissue are important factors in predicting obesity-associated diseases, but cellular microstructure of white adipose tissue has been less explored. To investigate the relationship between adipocyte size and obesity-related traits, and their underlying disease-driving genetic associations, we performed the largest study of automatic adipocyte phenotyping linking histological measurements and genetics to date. We introduce deep learning based methods for scalable and accurate semantic segmentation of subcutaneous and visceral adipose tissue histology samples (N=2,667) across 5 independent cohorts, including data from 9,000 whole slide images, with over 27 million adipocytes. Estimates of mean size of adipocytes were validated against Glastonbury et al. 2020. We show that adipocyte hypertrophy correlates with an adverse metabolic profile with increased levels of leptin, fasting plasma glucose, glycated hemoglobin and triglycerides, and decreased levels of adiponectin and HDL cholesterol. We performed the largest GWAS (N Subcutaneous = 2066, N Visceral = 1878) and subsequent meta-analysis of mean adipocyte area, and find two genome-wide significant loci (rs73184721, rs200047724) associated with increased 95%-quantile adipocyte size in respectively visceral and subcutaneous adipose tissue. Stratifying by sex, in females we find two genome-wide significant loci, with one variant (rs140503338) associated with increased mean adipocyte size in subcutaneous adipose tissue, and the other (rs11656704) is associated with decreased 95%-quantile adipocyte size in visceral adipose tissue.
Collapse
|
18
|
Krupka S, Aldehoff AS, Goerdeler C, Engelmann B, Rolle-Kampczyk U, Schubert K, Klöting N, von Bergen M, Blüher M. Metabolic and molecular Characterization, following dietary exposure to DINCH, Reveals new Implications for its role as a Metabolism-Disrupting chemical. ENVIRONMENT INTERNATIONAL 2025; 196:109306. [PMID: 39884247 DOI: 10.1016/j.envint.2025.109306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/20/2024] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
Plastic materials are ubiquitous, leading to constant human exposure to plastic additives such as plasticizers. There is growing evidence that plasticizers may contribute to obesity due to their disruptive effects on metabolism. Alternatives like diisononylcyclohexane-1,2-dicarboxylate (DINCH) are replacing traditional phthalates such as di-(2-ethylhexyl) phthalate (DEHP), which are now banned due to their proven harmful health effects. While DINCH is considered a safer alternative to DEHP and no adipogenic effects have been demonstrated in in vivo studies, recent research suggests that the primary metabolite, monoisononylcyclohexane-1,2-dicarboxylic acid ester (MINCH), promotes adipocyte differentiation and dysfunction in vitro. However, metabolic and molecular effects are not fully understood in vivo. Here, we performed a comprehensive in vivo analysis using C57BL/6N mice to investigate the effects of DINCH on adipose tissue physiology and function. Mice were exposed to two doses of DINCH for 16 weeks, followed by a 10-week recovery period. Tissue analysis confirmed the presence of DINCH and MINCH in liver and adipose tissue after treatment and recovery. After the recovery period, elevated DINCH concentrations in adipose tissue depots indicated possible bioaccumulation. Although no changes were observed in body composition and energy expenditure, sex-specific metabolic effects were identified. Female mice exhibited impaired whole-body insulin sensitivity and higher triglyceride levels, while male mice showed an altered insulin/C-peptide ratio and elevated cholesterol, HDL, and LDL levels. Proteomic profiling of serum, adipose and liver tissues revealed changes in pathways related to central energy metabolism and immune response, highlighting the systemic impact of DINCH, potentially on inflammatory processes. Most effects of DINCH, such as changes in insulin response and serum lipid levels, were diminished after the recovery period. Despite many findings consistent with the existing literature suggesting DINCH as a safer DEHP substitute, the observed sex-specific effects on insulin sensitivity, lipid metabolism and inflammatory processes, as well as potential bioaccumulation and long-term metabolic effects of DINCH exposure warrant careful consideration in further risk assessment.
Collapse
Affiliation(s)
- Sontje Krupka
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Centre München at the University of Leipzig Germany; Department of Endocrinology Nephrology Rheumatology University Hospital Leipzig Medical Research Center Leipzig Germany
| | - Alix Sarah Aldehoff
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany
| | - Cornelius Goerdeler
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany
| | - Beatrice Engelmann
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany
| | - Kristin Schubert
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany.
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Centre München at the University of Leipzig Germany.
| | - Martin von Bergen
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany; Institute of Biochemistry Faculty of Biosciences, Pharmacy and Psychology Leipzig University Leipzig Germany; German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig Leipzig Germany.
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Centre München at the University of Leipzig Germany; Department of Endocrinology Nephrology Rheumatology University Hospital Leipzig Medical Research Center Leipzig Germany
| |
Collapse
|
19
|
Han SM, Nahmgoong H, Yim KM, Kim JB. How obesity affects adipocyte turnover. Trends Endocrinol Metab 2025; 36:147-160. [PMID: 39095230 DOI: 10.1016/j.tem.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024]
Abstract
Cellular turnover is fundamental for tissue homeostasis and integrity. Adipocyte turnover, accounting for 4% of the total cellular mass turnover in humans, is essential for adipose tissue homeostasis during metabolic stress. In obesity, an altered adipose tissue microenvironment promotes adipocyte death. To clear dead adipocytes, macrophages are recruited and form a distinctive structure known as crown-like structure; subsequently, new adipocytes are generated from adipose stem and progenitor cells in the adipogenic niche to replace dead adipocytes. Accumulating evidence indicates that adipocyte death, clearance, and adipogenesis are sophisticatedly orchestrated during adipocyte turnover. In this Review, we summarize our current understandings of each step in adipocyte turnover, discussing its key players and regulatory mechanisms.
Collapse
Affiliation(s)
- Sang Mun Han
- National Leader Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hahn Nahmgoong
- National Leader Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung Min Yim
- National Leader Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae Bum Kim
- National Leader Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
20
|
Sa R, Zhang F, Zhang X, Gao W, Zhang Y, Gan J, Hou S, Gui L. Effects of different Lys/Met ratios on the antioxidant capacity, tissue morphology, and fatty acid composition of subcutaneous fat in Tibetan sheep on low-protein diets: a lipidomic analysis. Front Vet Sci 2025; 11:1528331. [PMID: 39949758 PMCID: PMC11824274 DOI: 10.3389/fvets.2024.1528331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 12/27/2024] [Indexed: 02/16/2025] Open
Abstract
Introduction This study employed lipidomics to investigate the effects of varying lysine (Lys)- to-methionine (Met) ratios on the antioxidant capacity, tissue morphology, and fatty acid composition of subcutaneous fat in Tibetan sheep fed a low-protein diet. Methods Ninety healthy male Tibetan sheep of similar body weight were randomly allocated into three groups. These sheep were fed a low-protein diet containing Lys/Met ratios of 1:1, 2:1, and 3:1. Ultra-High Performance Liquid Chromatography-tandem Mass Spectrometry (UHPLC-MS/MS) was employed to explore the changes in various lipid subclasses in subcutaneous adipose tissue. The expression of genes associated with adipogenesis, antioxidant capacity, and fatty acid metabolism was also examined. Results The results indicated that the 1:1 Lys/Met group exhibited significantly higher antioxidant capacity (glutathione peroxidase, GSH-Px), with more orderly adipocyte arrangement, uniform cell size, and a general increase in unsaturated fatty acid levels. Additionally, several lipid molecules associated with the phenotype (Antioxidant index and fatty acid content) were identified, namely, DG(38:3e) + Na, PE(17:1_22:2)-H, PI(17:0_20:3)-H, TG(33:0e) + NH4, Cer(d14:0_17:1) + H, and CL(81:13)-2H. Furthermore, the findings showed that the upregulation of PPARγ, FASN, FAD4, CPT1A, and GPX4 can enhance adipocyte differentiation and lipid accumulation, thereby improving metabolic function in subcutaneous adipose tissue via the regulation of lipid metabolism and oxidative defense mechanisms. Discussion In summary, this study provides a theoretical foundation for optimizing precision feeding strategies for Tibetan sheep, offering crucial data to support enhancements in production efficiency and meat quality.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Linsheng Gui
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, China
| |
Collapse
|
21
|
Xing Y, Ma C, Guan H, Shen J, Shen Y, Li G, Sun G, Tian Y, Kang X, Liu X, Li H, Tian W. Multi-Omics Insights into Regulatory Mechanisms Underlying Differential Deposition of Intramuscular and Abdominal Fat in Chickens. Biomolecules 2025; 15:134. [PMID: 39858528 PMCID: PMC11763713 DOI: 10.3390/biom15010134] [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: 12/11/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Excessive abdominal fat deposition in chickens disadvantages feed conversion, meat production, and reproductive performance. Intramuscular fat contributes to meat texture, tenderness, and flavor, serving as a vital indicator of overall meat quality. Therefore, a comprehensive analysis of the regulatory mechanisms governing differential deposition of abdominal versus intramuscular fat is essential in breeding higher-quality chickens with ideal fat distribution. This review systematically summarizes the regulatory mechanisms underlying intramuscular and abdominal fat traits at chromatin, genomic, transcriptional, post-transcriptional, translational, and epigenetic-modification scales. Additionally, we summarize the role of non-coding RNAs and protein-coding genes in governing intramuscular and abdominal fat deposition. These insights provide a valuable theoretical foundation for the genetic engineering of high-quality and high-yielding chicken breeds.
Collapse
Affiliation(s)
- Yuxin Xing
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
| | - Chenglin Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
| | - Hongbo Guan
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
| | - Jianing Shen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
| | - Ying Shen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
| | - Guoxi Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization of Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450046, China
| | - Guirong Sun
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization of Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450046, China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization of Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450046, China
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization of Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450046, China
| | - Xiaojun Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization of Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450046, China
| | - Hong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization of Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450046, China
| | - Weihua Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.X.); (C.M.); (H.G.); (J.S.); (Y.S.); (G.L.); (G.S.); (Y.T.); (X.K.); (X.L.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization of Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450046, China
| |
Collapse
|
22
|
Zhang FS, Li HJ, Yu X, Song YP, Ren YF, Qian XZ, Liu JL, Li WX, Huang YR, Gao K. Global trends and hotspots of type 2 diabetes in children and adolescents: A bibliometric study and visualization analysis. World J Diabetes 2025; 16:96032. [PMID: 39817223 PMCID: PMC11718446 DOI: 10.4239/wjd.v16.i1.96032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/30/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Epidemiological surveys indicate an increasing incidence of type 2 diabetes mellitus (T2DM) among children and adolescents worldwide. Due to rapid disease progression, severe long-term cardiorenal complications, a lack of effective treatment strategies, and substantial socioeconomic burdens, it has become an urgent public health issue that requires management and resolution. Adolescent T2DM differs from adult T2DM. Despite a significant increase in our understanding of youth-onset T2DM over the past two decades, the related review and evidence-based content remain limited. AIM To visualize the hotspots and trends in pediatric and adolescent T2DM research and to forecast their future research themes. METHODS This study utilized the terms "children", "adolescents", and "type 2 diabetes", retrieving relevant articles published between 1983 and 2023 from three citation databases within the Web of Science Core Collection (SCI, SSCI, ESCI). Utilizing CiteSpace and VoSviewer software, we analyze and visually represent the annual output of literature, countries involved, and participating institutions. This allows us to predict trends in this research field. Our analysis encompasses co-cited authors, journal overlays, citation overlays, time-zone views, keyword analysis, and reference analysis, etc. RESULTS A total of 9210 articles were included, and the annual publication volume in this field showed a steady growth trend. The United States had the highest number of publications and the highest H-index. The United States also had the most research institutions and the strongest research capacity. The global hot journals were primarily diabetes professional journals but also included journals related to nutrition, endocrinology, and metabolism. Keyword analysis showed that research related to endothelial dysfunction, exposure risk, cardiac metabolic risk, changes in gut microbiota, the impact on comorbidities and outcomes, etc., were emerging keywords. They have maintained their popularity in this field, suggesting that these areas have garnered significant research interest in recent years. CONCLUSION Pediatric and adolescent T2DM is increasingly drawing global attention, with genes, behaviors, environmental factors, and multisystemic interventions potentially emerging as future research hot spots.
Collapse
Affiliation(s)
- Fang-Shuo Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hai-Jing Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xue Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yi-Ping Song
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yan-Feng Ren
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xuan-Zhu Qian
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jia-Li Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wen-Xun Li
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yi-Ran Huang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Kuo Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
23
|
Wu Y, Deng S, Wei S, Wei W, He Y, Guo J. Adipocyte-Targeted Nanotechnology and Cell-Based Therapy for Obesity Treatment. ChemMedChem 2025; 20:e202400611. [PMID: 39390653 DOI: 10.1002/cmdc.202400611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/12/2024]
Abstract
Obesity is a critical risk factor for the development of metabolic diseases and is often associated with dysfunctional adipocytes. Prevalent treatments such as lifestyle intervention, pharmacotherapy, and bariatric surgery are often accompanied by adverse side effects and poor patient compliance. Nanotechnology and cell-based therapy offer innovative approaches for targeted obesity treatments, as they can directly target adipocytes, regulate lipid metabolism, and minimize off-target effects. Here, we provide an overview of the intricate relationship between adipocytes and obesity, highlighting the potential of nanotechnology and cell-based therapy in obesity treatment. Additionally, we discuss the advancements of adipose-derived mesenchymal stem cells (ADMSCs) in obesity progression, including the latest challenges and considerations for developing adipose-targeted treatments for obesity. The objective is to provide a perspective on the design and development of nanotechnology and cell-based therapy for treating obesity and related comorbidities.
Collapse
Affiliation(s)
- Yue Wu
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Siqi Deng
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Siyu Wei
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Wenqi Wei
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yunxiang He
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- Bioproducts Institute, Department of Chemical and Biological Engineering, The, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
- State Key Laboratory of Polymer Materials Engineering, Department of Chemical and Biological Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| |
Collapse
|
24
|
Wen X, Mao Y, Li Z, Chen G, Zhou S. Association between weight-adjusted waist index and Hashimoto's thyroiditis: insights from NHANES 2007-2012. Front Nutr 2025; 11:1520440. [PMID: 39834468 PMCID: PMC11743686 DOI: 10.3389/fnut.2024.1520440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
Objective While previous studies have explored the relationship between obesity and levels of thyroid autoantibodies, research using novel indicators such as weight-adjusted waist index (WWI) remains limited. This study aimed to evaluate the potential relationship between WWI and thyroid autoantibody levels, with the objective of improving our understanding of the links between central obesity and Hashimoto's thyroiditis (HT). Methods We conducted a cross-sectional study using data from the National Health and Nutrition Examination Survey (NHANES) cycles from 2007 to 2012. We analyzed the relationship between WWI and levels of thyroid peroxidase antibodies (TPOAb) and thyroglobulin antibodies (TgAb) through multivariate linear regression and subgroup analyses. Results The study included 7,056 participants with an average age of 49.71 ± 17.66 years, comprising 49.18% females. Mean WWI across the cohort was 11.04 ± 0.84. Analysis revealed a significant positive association between WWI and TPOAb levels (β: 4.78, 95% CI: 1.52, 8.05, p = 0.0041), which remained consistent across all multivariate linear regression models. In contrast, no significant correlation was found between WWI and TgAb levels after adjusting for covariates. Subgroup analysis stratified by gender demonstrated a notable gender-specific effect, where the positive correlation between WWI and TPOAb levels was evident only in females (β: 8.13, 95% CI: 4.14, 12.12, p < 0.0001). Conclusion This study used WWI as a novel indicator of central obesity and identified a strong association with HT, particularly notable in females. However, further high-quality studies are needed to confirm these findings and explore the underlying biological mechanisms.
Collapse
Affiliation(s)
- Xiaoyong Wen
- Department of Thyroid Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Mao
- Department of Thyroid Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zeyu Li
- Department of Thyroid Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guangji Chen
- Department of Surgery, University Hospital, Central South University, Changsha, Hunan, China
| | - Shiwei Zhou
- Department of Thyroid Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| |
Collapse
|
25
|
Yu X, Wang X, Dun S, Zhang H, Yao Y, Liu Z, Wang J, Liu W. Obesity modifies the association between abnormal glucose metabolism and atrial fibrillation in older adults: a community-based longitudinal and prospective cohort study. Hellenic J Cardiol 2025:S1109-9666(24)00270-7. [PMID: 39756654 DOI: 10.1016/j.hjc.2024.12.007] [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: 08/05/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025] Open
Abstract
OBJECTIVE To investigate the modifying role of obesity in the association between abnormal glucose metabolism and atrial fibrillation (AF) risk in older individuals. METHODS From April 2007 to November 2011, 11,663 participants aged ≥60 years were enrolled in the Shandong area. Glucose metabolic status was determined using fasting plasma glucose and hemoglobin A1c levels, and obesity was determined using body mass index (BMI), waist-to-hip ratio (WHR), and visceral fat area (VFA). Obesity-associated metabolic activities were assessed using the adiponectin-to-leptin ratio (ALR), galectin-3, and triglyceride-glucose index (TyG). New-onset AF was diagnosed by ICD-10. RESULTS During an average of 11.1 years of follow-up, 1343 participants developed AF. AF risks were higher in those with prediabetes, uncontrolled diabetes, and well-controlled diabetes than with normoglycemia. The hazard ratios were decreased by 14.79%, 40.29%, and 25.23% in those with prediabetes; 31.44%, 53.56%, and 41.90% in those with uncontrolled diabetes; and 21.16%, 42.38%, and 27.59% in those with well-controlled diabetes after adjusting for BMI, WHR, and VFA, respectively. The population-attributable risk percentages of general obesity, central obesity, and high VFA for new-onset AF were 10.43%, 34.78%, and 31.30%, respectively. ALR, galectin-3, and TyG significantly mediated the association of BMI, WHR, and VFA with AF risk (all Padj. < 0.001). CONCLUSION Obesity mediates the association between abnormal glucose metabolism and AF risk in older individuals. WHR is a more effective modifier than BMI and VFA for moderating the association. ALR, TyG, and galectin-3 mediate the moderating effect of obesity on the association between abnormal glucose metabolism and AF risk.
Collapse
Affiliation(s)
- Xinyi Yu
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China; Cardio-Cerebrovascular Control and Research Center, Clinical and Basic Medicine College, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xin Wang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Siyi Dun
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Hua Zhang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China; Cardio-Cerebrovascular Control and Research Center, Clinical and Basic Medicine College, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yanli Yao
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China; Cardio-Cerebrovascular Control and Research Center, Clinical and Basic Medicine College, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zhendong Liu
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China; Cardio-Cerebrovascular Control and Research Center, Clinical and Basic Medicine College, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| | - Juan Wang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250012, China.
| | - Weike Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
26
|
Fitzgibbons TP, Kogan S, Tran KV. Vascular-Adipose Crosstalk: Angiogenesis and Adipose Tissue Remodeling. Circ Res 2025; 136:112-114. [PMID: 39745990 PMCID: PMC11698489 DOI: 10.1161/circresaha.124.325899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Timothy P Fitzgibbons
- Division of Cardiovascular Medicine, Department of Medicine (T.P.F., K.-V.T.), University of Massachusetts Chan Medical School, Worcester, MA
| | - Sophia Kogan
- Department of Psychiatry (S.K.), University of Massachusetts Chan Medical School, Worcester, MA
| | - Khanh-Van Tran
- Division of Cardiovascular Medicine, Department of Medicine (T.P.F., K.-V.T.), University of Massachusetts Chan Medical School, Worcester, MA
| |
Collapse
|
27
|
Wang Z, Liu X, Sheng L, Xie Y, Feng W, Yu L. Effects of duration of high-fat diet on adipocyte hyperplasia in rat epididymis. Obes Res Clin Pract 2025; 19:54-62. [PMID: 39922761 DOI: 10.1016/j.orcp.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 06/25/2024] [Accepted: 02/01/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND High-fat diet (HFD) contributes to obesity and enhances the expression of mature adipocyte markers. However, the effect of HFD on adipocyte hyperplasia remains controversial. This may be due to variations in the duration of HFD. This study aimed to investigate the effects of different durations of HFD on adipocyte hyperplasia and the expression of mature adipocyte-related markers in obese rats. METHODS We divided 32 Sprague-Dawley rats into four groups: B (standard diet control), H1 (HFD for four weeks), H2 (HFD for eight weeks), and H3 (HFD for 12 weeks). We evaluated the morphological changes in epididymal fat cells, measured serum inflammatory markers using enzyme-linked immunosorbent assay (ELISA) kits, and quantified adipocyte hyperplasia and maturation markers using western blotting. RESULTS We observed progressive increases in body weight, epididymal fat weight, serum leptin, TNF-α, IL-6, irisin, PPARγ, adiponectin, and FNDC5 protein expression over 8 weeks of HFD. 12 weeks of HFD intervention resulted in significant decreases in irisin, PPARγ, adiponectin, and FNDC5. Concurrently, the expression of perilipin A and ATGL declined with prolonged HFD. CONCLUSIONS Our results suggest that the duration of HFD significantly affects adipocyte ability to undergo hyperplasia in the epididymis of obese rats. Specifically, 4 weeks of HFD did not change the capacity for adipocyte hyperplasia, while 8 weeks of the diet enhanced this capacity. Interestingly, a longer diet duration (12 weeks) led to a decrease in adipocyte hyperplasia.
Collapse
Affiliation(s)
- Zhaoxin Wang
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Xiujuan Liu
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China.
| | - Lei Sheng
- Department of Scientific Research, Nanjing Sport Institute, Nanjing, China.
| | - Yuting Xie
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Wanyu Feng
- Department of Science Experiment Center, Nanjing Sport Institute, Nanjing, China
| | - Li Yu
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| |
Collapse
|
28
|
Ko K, Bandara SR, Zhou W, Svenningsson L, Porras-Gómez M, Kambar N, Dreher-Threlkeld J, Topgaard D, Hernández-Saavedra D, Anakk S, Leal C. Diet-Induced Obesity Modulates Close-Packing of Triacylglycerols in Lipid Droplets of Adipose Tissue. J Am Chem Soc 2024; 146:34796-34810. [PMID: 39644234 DOI: 10.1021/jacs.4c13420] [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] [Indexed: 12/09/2024]
Abstract
Adipose-derived lipid droplets (LDs) are rich in triacylglycerols (TAGs), which regulate essential cellular processes, such as energy storage. Although TAG accumulation and LD expansion in adipocytes occur during obesity, how LDs dynamically package TAGs in response to excessive nutrients remains elusive. Here, we found that LD lipidomes display a remarkable increase in TAG acyl chain saturation under calorie-dense diets, turning them conducive to close-packing. Using high-resolution X-ray diffraction, solid-state NMR, and imaging, we show that beyond size expansion LDs from mice under varied obesogenic diets govern fat accumulation by packing TAGs in different crystalline polymorphs. Consistently, LDs and tissue stiffen for high-calorie-fed mice with more than a 2-fold increase in elastic moduli compared to normal diet. Our data suggest that in addition to expanding, adipocyte LDs undergo structural remodeling by close-packing rigid and highly saturated TAGs in response to caloric overload, as opposed to liquid TAGs in a low-calorie diet. This work provides insights into how lipid packing within LDs can allow for the rapid and optimal expansion of fat during the initial stages of obesity.
Collapse
Affiliation(s)
- Kyungwon Ko
- Department of Bioengineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Sarith R Bandara
- Department of Materials Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Weinan Zhou
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Leo Svenningsson
- Division of Physical Chemistry, Lund University, Lund 22100, Sweden
| | - Marilyn Porras-Gómez
- Department of Materials Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Nurila Kambar
- Department of Materials Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Julia Dreher-Threlkeld
- Department of Materials Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Daniel Topgaard
- Division of Physical Chemistry, Lund University, Lund 22100, Sweden
| | - Diego Hernández-Saavedra
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Sayeepriyadarshini Anakk
- Department of Bioengineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Cecília Leal
- Department of Bioengineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Materials Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
29
|
Griesel BA, Olson AL. PFKFB3 protein in adipose tissue contributes to whole body glucose homeostasis. FASEB J 2024; 38:e70254. [PMID: 39659238 DOI: 10.1096/fj.202402070r] [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: 09/03/2024] [Revised: 11/19/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Age-dependent changes in adipose tissue are thought to play a role in development of insulin resistance. A major age-dependent change in adipose tissue is the downregulation of key proteins involved in carbohydrate metabolism. In the current study, we investigate the role of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) a key governor of the rate of glycolysis in adipocytes via the synthesis of fructose-2,6-bisphosphate that was significantly downregulated in aged mice. We employed an adipocyte-specific PFKFB3 mouse line to investigate the role of PFKFB3 on adipocyte function. In both aged mice and PFKFB3-knockout mice, we observed an increase in O-glcNAcylated proteins consistent with a shift in glucose metabolism toward the hexosamine biosynthetic pathway. Under chow-fed conditions, PFKFB3 knockout resulted in significantly smaller adipocyte area, but no difference in total fat mass. While glucose tolerance was unchanged under chow conditions, when mice were challenged with a 4 weeks high-fat feeding, PFKFB3 deletion led to a greater decrease in glucose tolerance as well as a significant increase in macrophage infiltration. These results indicate that perturbation of the glycolytic pathway in adipose tissue has multiple effects of adipocyte biology and may play a significant role in metabolic changes associated with aging. Results of this student support the notion that changes in glucose metabolism in adipose tissue impact whole-body metabolism.
Collapse
Affiliation(s)
- Beth A Griesel
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - Ann Louise Olson
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| |
Collapse
|
30
|
Tambasco D, Tomaselli F, Albanese R. Effects of Different Bariatric Procedures in Histological Changes in Skin and Subcutaneous Cellular Tissue in Massive Weight Loss Patients. Aesthetic Plast Surg 2024:10.1007/s00266-024-04600-2. [PMID: 39658668 DOI: 10.1007/s00266-024-04600-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024]
Affiliation(s)
- Damiano Tambasco
- Plastic Surgery Unit, San Carlo di Nancy Hospital, Via Aurelia 275, Rome, Italy
| | - Federica Tomaselli
- Plastic Surgery Unit, San Carlo di Nancy Hospital, Via Aurelia 275, Rome, Italy
| | - Roberta Albanese
- Plastic Surgery Unit, San Carlo di Nancy Hospital, Via Aurelia 275, Rome, Italy.
- Clinic of Plastic and Reconstructive Surgery, Department of Medical Area (DAME), Academic Hospital of Udine, University of Udine, Piazzale Santa Maria della Misericordia 15, 33100, Udine, Italy.
- Santa Maria Misericordia Hospital, Piazzale Santa Maria della Misericordia, 15, 33100, Udine, UD, Italy.
| |
Collapse
|
31
|
Kitaghenda FK, Wang J, Li T, Hong J, Yao L, Zhu X. Normalization of WISP1 circulating level and tissue expression following metabolic and bariatric surgery using rat model. Updates Surg 2024; 76:2841-2849. [PMID: 39407056 DOI: 10.1007/s13304-024-01977-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/30/2024] [Indexed: 12/10/2024]
Abstract
Wingless-type inducible signaling pathway protein-1 (WISP1) is a newly recognized adipokine, associated with obesity and type 2 diabetes (T2DM). This study aimed to investigate the effect of metabolic and bariatric surgery (MBS) on WISP1 circulating (serum) levels and tissue expression using rat models. We initially investigated whether WISP1 circulating levels were altered between the T2DM and normal rats. After confirmation, Sprague-Dawley (SD) rats were obtained and randomly divided as follows: Roux-en-Y gastric bypass (RYGB) group (n = 10), sleeve gastrectomy (SG) group (n = 10), SHAM group (n = 10), and normal control (NC) group (n = 10). Rats were followed for 8 weeks postoperatively. Preoperative and postoperative WISP1 circulating (serum) levels, glucose tolerance test (OGTT), insulin tolerance test (ITT), postoperative WISP1 expression (visceral adipose tissue, VAT; and skeletal muscle, SM), body weight, food intake, and fasting blood glucose levels were recorded. MBS significantly induced glucose control and weight loss. At postoperative week 8, WISP1 serum levels decreased in the MBS groups (P < 0.05); furthermore, WISP1 expression in VAT and SM significantly decreased in the RYGB and SG groups than SHAM (P < 0.05, and P < 0.05, respectively). Whereas the difference in the expression level between SG and RYGB did not amount to statistical significance (P > 0.05). MBS significantly decreased WISP1 serum levels, tissue expression in the VAT, and SM. As WISP1 is a regulator of low-grade inflammation associated with obesity and T2DM, further studies are needed to explore its relevance in MBS.
Collapse
Affiliation(s)
- Fidele Kakule Kitaghenda
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Jian Wang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Tianci Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Jian Hong
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Libin Yao
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.
| | - Xiaocheng Zhu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
32
|
Yagi M, Sakai A, Yasutomi S, Suzuki K, Kashikura H, Goto K. Assessment of Tail-Cutting in Frozen Albacore ( Thunnus alalunga) Through Ultrasound Inspection and Chemical Analysis. Foods 2024; 13:3860. [PMID: 39682932 DOI: 10.3390/foods13233860] [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: 10/18/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Fat content is the main criterion for evaluating albacore quality. However, no reports exist on the accuracy of the tail-cutting method, a method used to assess the fat content of albacore. Here, we evaluated this method by comparing it with chemical analysis and ultrasound inspection. We measured the actual fat content in albacore using chemical analysis and compared the results with those obtained using the tail-cutting method. Significant discrepancies (99% CI, t-test) were observed in fat content among the tail-cutting samples. Using chemical analysis as the ground truth, the accuracy of tail-cutting from two different companies was 70.0% for company A and 51.9% for company B. An ultrasound inspection revealed that a higher fat content reduced the amplitude of ultrasound signals with statistical significance (99% CI, t-test). Finally, machine learning algorithms were used to enforce the ultrasound inspection. The best combination of ultrasound inspection and a machine learning algorithm achieved an 84.2% accuracy for selecting fat-rich albacore, which is better than tail-cutting (73.6%). Our findings suggested that ultrasound inspection could be a valuable and non-destructive method for estimating the fat content of albacore, achieving better accuracy than the traditional tail-cutting method.
Collapse
Affiliation(s)
- Masafumi Yagi
- School of Marine Science and Technology, Tokai University, 3-20-1 Orido, Shimizu-ku, Shizuoka-shi 424-8610, Shizuoka, Japan
| | - Akira Sakai
- Artificial Intelligence Laboratory, Fujitsu Limited, 4-1-1 Kamikodanaka, Nakahara-ku, Kawasaki-shi 211-8588, Kanagawa, Japan
| | - Suguru Yasutomi
- Artificial Intelligence Laboratory, Fujitsu Limited, 4-1-1 Kamikodanaka, Nakahara-ku, Kawasaki-shi 211-8588, Kanagawa, Japan
| | - Kanata Suzuki
- Artificial Intelligence Laboratory, Fujitsu Limited, 4-1-1 Kamikodanaka, Nakahara-ku, Kawasaki-shi 211-8588, Kanagawa, Japan
| | - Hiroki Kashikura
- Graduate School of Marine Science and Technology, Tokai University, 3-20-1 Orido, Shimizu-ku, Shizuoka-shi 424-8610, Shizuoka, Japan
| | - Keiichi Goto
- School of Marine Science and Technology, Tokai University, 3-20-1 Orido, Shimizu-ku, Shizuoka-shi 424-8610, Shizuoka, Japan
| |
Collapse
|
33
|
Malicka A, Ali A, MacCannell ADV, Roberts LD. Brown and beige adipose tissue-derived metabokine and lipokine inter-organ signalling in health and disease. Exp Physiol 2024. [PMID: 39591977 DOI: 10.1113/ep092008] [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: 07/19/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024]
Abstract
Adipose tissue has an established endocrine function through the secretion of adipokines. However, a role for bioactive metabolites and lipids, termed metabokines and lipokines, is emerging in adipose tissue-mediated autocrine, paracrine and endocrine signalling and inter-organ communication. Traditionally seen as passive entities, metabolites are now recognized for their active roles in regulating cellular signalling and local and systemic metabolism. Distinct from white adipose tissue, specific endocrine functions have been attributed to thermogenic brown and beige adipose tissues. Brown and beige adipose tissues have been identified as sources of metabokines and lipokines, which influence diverse metabolic pathways, such as fatty acid β-oxidation, mitochondrial function and glucose homeostasis, across a range of tissues, including skeletal muscle, adipose tissue and heart. This review explores the intricate signalling mechanisms of brown and beige adipose tissue-derived metabokines and lipokines, emphasizing their roles in maintaining metabolic homeostasis and their potential dysregulation in metabolic diseases. Furthermore, we discuss the therapeutic potential of targeting these pathways, proposing that precise modulation of metabokine receptors and transporters could offer superior specificity and efficacy in comparison to conventional approaches, such as β-adrenergic signalling-stimulated activation of brown adipose tissue thermogenesis. Understanding the complex interactions between adipokines, metabokines and lipokines is essential for developing a systems-level approach to new interventions for metabolic disorders, underscoring the need for continued research in this rapidly evolving field.
Collapse
Affiliation(s)
- Anna Malicka
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Aysha Ali
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Amanda D V MacCannell
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
34
|
Ryskova L, Pospisilova K, Vavra J, Wolf T, Dvorak A, Vitek L, Polak J. Contribution of glucose and glutamine to hypoxia-induced lipid synthesis decreases, while contribution of acetate increases, during 3T3-L1 differentiation. Sci Rep 2024; 14:28193. [PMID: 39548264 PMCID: PMC11568125 DOI: 10.1038/s41598-024-79458-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/08/2024] [Indexed: 11/17/2024] Open
Abstract
The molecular mechanisms linking obstructive sleep apnea syndrome (OSA) to obesity and the development of metabolic diseases are still poorly understood. The role of hypoxia (a characteristic feature of OSA) in excessive fat accumulation has been proposed. The present study investigated the possible effects of hypoxia (4% oxygen) on de novo lipogenesis by tracking the major carbon sources in differentiating 3T3-L1 adipocytes. Gas-permeable cultuware was employed to cultivate 3T3-L1 adipocytes in hypoxia (4%) for 7 or 14 days of differentiation. We investigated the contribution of glutamine, glucose or acetate using 13C or 14C labelled carbons to the newly synthesized lipid pool, changes in intracellular lipid content after inhibiting citrate- or acetate-dependent pathways and gene expression of involved key enzymes. The results demonstrate that, in differentiating adipocytes, hypoxia decreased the synthesis of lipids from glucose (44.1 ± 8.8 to 27.5 ± 3.0 pmol/mg of protein, p < 0.01) and partially decreased the contribution of glutamine metabolized through the reverse tricarboxylic acid cycle (4.6% ± 0.2-4.2% ± 0.1%, p < 0.01). Conversely, the contribution of acetate, a citrate- and mitochondria-independent source of carbons, increased upon hypoxia (356.5 ± 71.4 to 649.8 ± 117.5 pmol/mg of protein, p < 0.01). Further, inhibiting the citrate- or acetate-dependent pathways decreased the intracellular lipid content by 58% and 73%, respectively (p < 0.01) showing the importance of de novo lipogenesis in hypoxia-exposed adipocytes. Altogether, hypoxia modified the utilization of carbon sources, leading to alterations in de novo lipogenesis in differentiating adipocytes and increased intracellular lipid content.
Collapse
Affiliation(s)
- Lucie Ryskova
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Ruska 87, Prague, 100 00, Czech Republic
| | - Katerina Pospisilova
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jiri Vavra
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Tomas Wolf
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Ruska 87, Prague, 100 00, Czech Republic
| | - Ales Dvorak
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Libor Vitek
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Internal Medicine, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital in Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Polak
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Ruska 87, Prague, 100 00, Czech Republic.
- Department of Internal Medicine, Thomayer University Hospital, Videnska 800, Prague, 140 59, Czech Republic.
| |
Collapse
|
35
|
Plissonneau C, Santosa S. Regional primary preadipocyte characteristics in humans with obesity and type 2 diabetes mellitus. Heliyon 2024; 10:e39710. [PMID: 39553621 PMCID: PMC11564010 DOI: 10.1016/j.heliyon.2024.e39710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
The excessive accumulation of adipose tissue in obesity appears to result in adipose tissue dysfunction perpetuating the onset of obesity-related diseases, including type 2 diabetes (T2DM). In humans, adipose tissue is stored in several depots including subcutaneous and visceral. These depots contribute to the pathology of obesity differently owing to differences in the tissue microenvironment, a main one being preadipocyte function. In examining adipocyte and preadipocyte characteristics, many have used the 3T3-L1 murine cell lines. Though these cell lines provide valuable mechanistic data, the results remain to be translated to humans. Experiments using primary human preadipocytes has shown that obesity and T2DM impact preadipocyte phenotypes. The objective of this review is to describe the differences in regional characteristics of primary preadipocytes collected from humans with obesity and to discuss how these characteristics might be affected in type 2 diabetes mellitus. In doing so, we will show that the characteristics of regional primary preadipocytes in humans are differentially affected by obesity and the development of T2DM.
Collapse
Affiliation(s)
- Claire Plissonneau
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Montreal, Quebec, Canada
- Metabolism, Obesity, and Nutrition Lab, School of Health, Concordia University, Montreal, Quebec, Canada
- Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| | - Sylvia Santosa
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Montreal, Quebec, Canada
- Metabolism, Obesity, and Nutrition Lab, School of Health, Concordia University, Montreal, Quebec, Canada
- Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| |
Collapse
|
36
|
Çil EN, Soysal Y. Anti-Obesity Effects of Calcium Fructoborate by Inhibiting Adipogenesis and Increasing SIRT's Expression in 3T3-L1 Cells. Biol Trace Elem Res 2024:10.1007/s12011-024-04444-6. [PMID: 39531139 DOI: 10.1007/s12011-024-04444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Obesity is a global public health problem that can lead to mortality and morbidity. Studies on the pathophysiology of obesity for effective and safe treatments are focused on the mechanisms of adipogenesis. The association between boron treatment and weight loss has been reported, but its anti-adipogenic mechanisms and effects on preadipocytes remain unclear. This study aims to investigate the effects of boron compounds boric acid (BA) and calcium fructoborate (CaFB) on adipogenesis using the most widely used in vitro 3T3-L1 cellular model. In our study, cytotoxicity, Oil Red O (ORO), gene and protein expression analyses and cellular NAD measurements of boron compounds were performed. Peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer binding protein α (C/EBPα) transcription factors are the main regulators of adipogenesis, and boron compounds affect them at gene and protein levels by showing anti-obesity effects. This is the first study to show that CaFB has anti-obesity properties in mouse adipocytes. Sirtuins, known as the longevity genes, were also activated from boron treatment. Results of this research provide new basic knowledge and insights into the effect of boron-based compounds on obesity. It also offers potential prospects for the development of effective treatment and/or supportive treatment methods.
Collapse
Affiliation(s)
- Ezgi Nur Çil
- Department of Molecular Medicine, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey.
| | - Yasemin Soysal
- Department of Molecular Medicine, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
37
|
Ahmed B, Farb MG, Gokce N. Cardiometabolic implications of adipose tissue aging. Obes Rev 2024; 25:e13806. [PMID: 39076025 DOI: 10.1111/obr.13806] [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: 08/21/2023] [Revised: 03/14/2024] [Accepted: 07/05/2024] [Indexed: 07/31/2024]
Abstract
Adipose tissue is a large endocrine organ that serves numerous physiological functions. As we age, adipose tissue remodels and can develop functional changes that alters its phenotype, potentially contributing to metabolic and cardiovascular disorders. Aging adipose tissue is characterized by regional redistribution of fat, accumulation of senescent cells, fibrosis, and decline in adipocyte differentiation capacities, which collectively impact adipose tissue function and whole body health. A notable transformation involves increased accumulation of intra-abdominal visceral adipose tissue and ectopic fat around internal organs such as the heart, blood vessels, liver, and kidneys that alter their functions. Other changes associated with aging include alterations in adipokine secretion and changes in adipocyte size and numbers. Aging adipocytes play a role in mediating chronic inflammation, metabolic dysfunction, and insulin resistance. Visceral adipose tissue, which increases in volume with aging, is in particular associated with inflammation, angiogenic dysfunction, and microvascular abnormalities, and mediators released by visceral fat may have adverse consequences systemically in multiple target organs, including the cardiovascular system. Understanding mechanisms underlying adipose tissue aging and its impact on cardiovascular health are important for developing interventions and treatments to promote healthy aging and reduce cardiometabolic disease risk.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Melissa G Farb
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
38
|
Deng L, Huang Y, Zhao F, Chen P, Huang X. Lack of adipocyte FAM20C improves whole body glucose homeostasis. Physiol Rep 2024; 12:e70126. [PMID: 39532808 PMCID: PMC11557440 DOI: 10.14814/phy2.70126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
FAM20C, a member of the family with sequence similarity 20, is involved in many physiological functions. Obesity, characterized by excessive accumulation of adipose tissue, has attracted more and more attention as a worldwide health problem. Here we generated adipocyte-specific FAM20C knockout mice to investigate the role of FAM20C in adipose tissue expansion and obesity. Our results demonstrate that knockout mice are protected against high fat diet-induced obesity, adiposity, and fatty liver disease. Additionally, knockout mice exhibited improved metabolic phenotypes, including enhanced glucose tolerance and insulin sensitivity compared with control mice. Furthermore, we observed reduced inflammatory infiltration and collagen deposition in the adipose tissues of knockout mice. Taken together, our results indicate that targeting FAM20C in adipocytes may be a promising strategy for the treatment of obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Liping Deng
- Department of EndocrinologyLonggang District Central Hospital of ShenzhenShenzhenGuangdongChina
| | - Yanshan Huang
- Division of Preventive HealthLonggang District Central Hospital of ShenzhenShenzhenGuangdongChina
- Department of NursingSchool of Medicine, Shantou UniversityShantouGuangdongChina
| | - Feifei Zhao
- Department of NursingShenzhen Clinical Medical College, Guangzhou University of Chinese MedicineShenzhenGuangdongChina
| | - Puxin Chen
- Department of EndocrinologyLonggang District Central Hospital of ShenzhenShenzhenGuangdongChina
| | - Xiaohong Huang
- Department of NursingLonggang District Central Hospital of ShenzhenShenzhenGuangdongChina
| |
Collapse
|
39
|
Tol MJ, Shimanaka Y, Bedard AH, Sapia J, Cui L, Colaço-Gaspar M, Hofer P, Ferrari A, Qian K, Kennelly JP, Lee SD, Gao Y, Xiao X, Gao J, Mack JJ, Weston TA, Pan C, Lusis AJ, Williams KJ, Su B, Pike DP, Reed A, Milosevich N, Cravatt BF, Arita M, Young SG, Ford DA, Zechner R, Vanni S, Tontonoz P. Dietary control of peripheral adipose storage capacity through membrane lipid remodelling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620374. [PMID: 39554041 PMCID: PMC11565995 DOI: 10.1101/2024.10.25.620374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Complex genetic and dietary cues contribute to the development of obesity, but how these are integrated on a molecular level is incompletely understood. Here, we show that PPARγ supports hypertrophic expansion of adipose tissue via transcriptional control of LPCAT3, a membrane-bound O-acyltransferase that enriches diet-derived omega-6 ( n -6) polyunsaturated fatty acids (PUFAs) in the phospholipidome. In high-fat diet-fed mice, lowering membrane n -6 PUFA levels by adipocyte-specific Lpcat3 knockout ( Lpcat3 AKO ) or by dietary lipid manipulation leads to dysfunctional triglyceride (TG) storage, ectopic fat deposition and insulin resistance. Aberrant lipolysis of stored TGs in Lpcat3 AKO adipose tissues instigates a non-canonical adaptive response that engages a futile lipid cycle to increase energy expenditure and limit further body weight gain. Mechanistically, we find that adipocyte LPCAT3 activity promotes TG storage by selectively enriching n -6 arachidonoyl-phosphatidylethanolamine at the ER-lipid droplet interface, which in turn favours the budding of large droplets that exhibit greater resistance to ATGL-dependent hydrolysis. Thus, our study highlights the PPARγ-LPCAT3 pathway as a molecular link between dietary n -6 PUFA intake, adipose expandability and systemic energy balance.
Collapse
|
40
|
Neeland IJ, Lim S, Tchernof A, Gastaldelli A, Rangaswami J, Ndumele CE, Powell-Wiley TM, Després JP. Metabolic syndrome. Nat Rev Dis Primers 2024; 10:77. [PMID: 39420195 DOI: 10.1038/s41572-024-00563-5] [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: 09/17/2024] [Indexed: 10/19/2024]
Abstract
The metabolic syndrome (MetS) is a multiplex modifiable risk factor for cardiovascular disease, type 2 diabetes mellitus and other health outcomes, and is a major challenge to clinical practice and public health. The rising global prevalence of MetS, driven by urbanization, sedentary lifestyles and dietary changes, underlines the urgency of addressing this syndrome. We explore the complex underlying mechanisms, including genetic predisposition, insulin resistance, accumulation of dysfunctional adipose tissue and ectopic lipids in abdominal obesity, systemic inflammation and dyslipidaemia, and how they contribute to the clinical manifestations of MetS. Diagnostic approaches vary but commonly focus on abdominal obesity (assessed using waist circumference), hyperglycaemia, dyslipidaemia and hypertension, highlighting the need for population-specific and phenotype-specific diagnostic strategies. Management of MetS prioritizes lifestyle modifications, such as healthy dietary patterns, physical activity and management of excess visceral and ectopic adiposity, as foundational interventions. We also discuss emerging therapies, including new pharmacological treatments and surgical options, providing a forward-looking perspective on MetS research and care. This Primer aims to inform clinicians, researchers and policymakers about MetS complexities, advocating for a cohesive, patient-centred management and prevention strategy. Emphasizing the multifactorial nature of MetS, this Primer calls for integrated public health efforts, personalized care and innovative research to address this escalating health issue.
Collapse
Affiliation(s)
- Ian J Neeland
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Division of Cardiovascular Medicine, University Hospitals Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.
| | - André Tchernof
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, Québec, Canada
| | - Amalia Gastaldelli
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Janani Rangaswami
- Division of Nephrology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Chiadi E Ndumele
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tiffany M Powell-Wiley
- Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Intramural Research Program, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Jean-Pierre Després
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, Québec, Canada.
- VITAM - Centre de recherche en santé durable, Centre intégré universitaire de santé et de services sociaux de la Capitale-Nationale, Québec, Québec, Canada.
| |
Collapse
|
41
|
Brito ML, Coutinho-Wolino KS, Almeida PP, Trigueira PDC, Alves APDP, Magliano DC, Stockler-Pinto MB. Unstressing the Reticulum: Nutritional Strategies for Modulating Endoplasmic Reticulum Stress in Obesity. Mol Nutr Food Res 2024; 68:e2400361. [PMID: 39363792 DOI: 10.1002/mnfr.202400361] [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: 05/15/2024] [Revised: 09/03/2024] [Indexed: 10/05/2024]
Abstract
The progression of obesity involves several molecular mechanisms that are closely associated with the pathophysiological response of the disease. Endoplasmic reticulum (ER) stress is one such factor. Lipotoxicity disrupts endoplasmic reticulum homeostasis in the context of obesity. Furthermore, it induces ER stress by activating several signaling pathways via inflammatory responses and oxidative stress. ER performs crucial functions in protein synthesis and lipid metabolism; thus, triggers such as lipotoxicity can promote the accumulation of misfolded proteins in the organelle. The accumulation of these proteins can lead to metabolic disorders and chronic inflammation, resulting in cell death. Thus, alternatives, such as flavonoids, amino acids, and polyphenols that are associated with antioxidant and anti-inflammatory responses have been proposed to attenuate this response by modulating ER stress via the administration of nutrients and bioactive compounds. Decreasing inflammation and oxidative stress can reduce the expression of several ER stress markers and improve clinical outcomes through the management of obesity, including the control of body weight, visceral fat, and lipid accumulation. This review explores the metabolic changes resulting from ER stress and discusses the role of nutritional interventions in modulating the ER stress pathway in obesity.
Collapse
Affiliation(s)
- Michele Lima Brito
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | - Karen Salve Coutinho-Wolino
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | - Patricia Pereira Almeida
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | | | - Ana Paula de Paula Alves
- Endocrinology Post Graduate Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 24210-201, Brazil
| | - D'Angelo Carlo Magliano
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Endocrinology Post Graduate Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 24210-201, Brazil
- Morphology Department, Biomedical Institute, Fluminense Federal University (UFF), Niterói, RJ, 24020-150, Brazil
| | - Milena Barcza Stockler-Pinto
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Nutrition Sciences Postgraduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24020-140, Brazil
| |
Collapse
|
42
|
Stanek E, Czamara K, Kaczor A. Increased obesogenic action of palmitic acid during early stage of adipogenesis. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159525. [PMID: 38876269 DOI: 10.1016/j.bbalip.2024.159525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/18/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
The functional differences between preadipocytes and fully differentiated mature adipocytes derived from stromal vascular fraction stem cells, as well as primary adipocytes have been analysed by evaluating their response to the obesogenic factor (a saturated fatty acid) and TNF-triggered inflammation. The analysis of single adipocytes shows that the saturated fatty acid (palmitic acid) accumulation is accompanied by inflammation and considerably dependent on the stage of the adipogenesis. In particular, preadipocytes show the exceptional potential for palmitic acid uptake resulting in their hypertrophy and the elevated cellular expression of the inflammation marker (ICAM-1). Our research provides new information on the impact of obesogenic factors on preadipocytes that is important in the light of childhood obesity prevention.
Collapse
Affiliation(s)
- Ewa Stanek
- Jagiellonian University, Doctoral School of Exact and Natural Sciences, 11 Lojasiewicza Str., 30-348 Krakow, Poland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Krzysztof Czamara
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), 14 Bobrzynskiego Str., 30-348 Krakow, Poland.
| | - Agnieszka Kaczor
- Jagiellonian University, Faculty of Chemistry, 2 Gronostajowa Str., 30-387 Krakow, Poland.
| |
Collapse
|
43
|
Di Rocco G, Trivisonno A, Trivisonno G, Toietta G. Dissecting human adipose tissue heterogeneity using single-cell omics technologies. Stem Cell Res Ther 2024; 15:322. [PMID: 39334440 PMCID: PMC11437900 DOI: 10.1186/s13287-024-03931-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Single-cell omics technologies that profile genes (genomic and epigenomic) and determine the abundance of mRNA (transcriptomic), protein (proteomic and secretomic), lipids (lipidomic), and extracellular matrix (matrisomic) support the dissection of adipose tissue heterogeneity at unprecedented resolution in a temporally and spatially defined manner. In particular, cell omics technologies may provide innovative biomarkers for the identification of rare specific progenitor cell subpopulations, assess transcriptional and proteomic changes affecting cell proliferation and immunomodulatory potential, and accurately define the lineage hierarchy and differentiation status of progenitor cells. Unraveling adipose tissue complexity may also provide for the precise assessment of a dysfunctional state, which has been associated with cancer, as cancer-associated adipocytes play an important role in shaping the tumor microenvironment supporting tumor progression and metastasis, obesity, metabolic syndrome, and type 2 diabetes mellitus. The information collected by single-cell omics has relevant implications for regenerative medicine because adipose tissue is an accessible source of multipotent cells; alternative cell-free approaches, including the use of adipose tissue stromal cell-conditioned medium, extracellular vesicles, or decellularized extracellular matrix, are clinically valid options. Subcutaneous white adipose tissue, which is generally harvested via liposuction, is highly heterogeneous because of intrinsic biological variability and extrinsic inconsistencies in the harvesting and processing procedures. The current limited understanding of adipose tissue heterogeneity impinges on the definition of quality standards appropriate for clinical translation, which requires consistency and uniformity of the administered product. We review the methods used for dissecting adipose tissue heterogeneity and provide an overview of advances in omics technology that may contribute to the exploration of heterogeneity and dynamics of adipose tissue at the single-cell level.
Collapse
Affiliation(s)
- Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Angelo Trivisonno
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | | | - Gabriele Toietta
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi, 53, 00144, Rome, Italy.
| |
Collapse
|
44
|
Ahn C, Divoux A, Zhou M, Seldin MM, Sparks LM, Whytock KL. An optimized pipeline for high-throughput bulk RNA-Seq deconvolution illustrates the impact of obesity and weight loss on cell composition of human adipose tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614489. [PMID: 39386599 PMCID: PMC11463495 DOI: 10.1101/2024.09.23.614489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Cellular heterogeneity of human adipose tissue, is linked to the pathophysiology of obesity and may impact the response to energy restriction and changes in fat mass. Here, we provide an optimized pipeline to estimate cellular composition in human abdominal subcutaneous adipose tissue (ASAT) from publicly available bulk RNA-Seq using signature profiles from our previously published full-length single nuclei (sn)RNA-Seq of the same depot. Individuals with obesity had greater proportions of macrophages and lower proportions of adipocyte sub-populations and vascular cells compared with lean individuals. Two months of diet-induced weight loss (DIWL) increased the estimated proportions of macrophages; however, two years of DIWL reduced the estimated proportions of macrophages, thereby suggesting a bi-phasic nature of cellular remodeling of ASAT during weight loss. Our optimized high-throughput pipeline facilitates the assessment of composition changes of highly characterized cell types in large numbers of ASAT samples using low-cost bulk RNA-Seq. Our data reveal novel changes in cellular heterogeneity and its association with cardiometabolic health in humans with obesity and following weight loss.
Collapse
Affiliation(s)
- Cheehoon Ahn
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Adeline Divoux
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Mingqi Zhou
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA
| | - Marcus M Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| |
Collapse
|
45
|
Koutaki D, Paltoglou G, Manou M, Vourdoumpa A, Ramouzi E, Tzounakou AM, Michos A, Bacopoulou F, Mantzou E, Zoumakis E, Papadopoulou M, Kassari P, Charmandari E. The Role of Secreted Frizzled-Related Protein 5 (Sfrp5) in Overweight and Obesity in Childhood and Adolescence. Nutrients 2024; 16:3133. [PMID: 39339733 PMCID: PMC11434931 DOI: 10.3390/nu16183133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objective: Secreted frizzled-related protein 5 (Sfrp5) is an anti-inflammatory adipokine that has been implicated in the pathophysiology of obesity and its metabolic complications. Despite the fact that numerous studies have been carried out in adults, limited data on Sfrp5 exist for youth, especially in relation to overweight and obesity. Methods: In our study, we assessed the concentrations of Sfrp5, total oxidative (TOS) and antioxidative (TAS) status, high-sensitivity C-reactive protein (hs-CRP), and several cytokines (IL-1α, IL-1β, IL-2, IL-6, IL-8, IL-12, TNF-α) in 120 children and adolescents (mean age ± SE: 11.48 ± 0.25 years; 48 prepubertal, 72 pubertal; 74 males and 46 females) before and 1 year after the implementation of a personalized, structured, lifestyle intervention program of healthy diet, sleep, and physical exercise. Results: Based on the body mass index (BMI), participants were categorized as having morbid obesity (n = 63, 52.5%), obesity (n = 21, 17.5%), overweight (n = 22, 18.33%), or normal BMIs (n = 14, 11.67%), based on the International Obesity Task Force (IOTF) cut-off points. Following the 1-year lifestyle intervention program, a significant improvement in anthropometric measurements (BMI, BMI-z score, diastolic blood pressure, WHR, and WHtR), body-composition parameters, hepatic enzymes, lipid profile, inflammation markers, and the insulin-sensitivity profile (HbA1C, HOMA index) was observed in all subjects. Sfrp5 decreased in subjects with obesity (p < 0.01); however, it increased significantly (p < 0.05) in patients with morbid obesity. Linear regression analysis indicates that TNF-α and systolic blood pressure were the best positive predictors and hs-CRP was the best negative predictor for Sfpr5 concentration at initial assessment and glucose concentration for ΔSfrp5, while TNF-α and TAS were the best positive predictors for Sfpr5 concentration at annual assessment. Conclusions: These results indicate that Sfrp5 is associated with severe obesity and is increased following weight loss in children and adolescents with morbid obesity. It is also related to metabolic homeostasis, as well as inflammation and oxidative status.
Collapse
Affiliation(s)
- Diamanto Koutaki
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - George Paltoglou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Maria Manou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
- Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Aikaterini Vourdoumpa
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Eleni Ramouzi
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Anastasia-Maria Tzounakou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Athanasios Michos
- Division of Infectious Diseases, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Flora Bacopoulou
- University Research Institute of Maternal and Child Health and Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Emilia Mantzou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Emmanouil Zoumakis
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Marina Papadopoulou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Penio Kassari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
46
|
Zhao C, Wen Z, Gao Y, Xiao F, Yan J, Wang X, Meng T. Pantothenic Acid Alleviates Fat Deposition and Inflammation by Suppressing the JNK/P38 MAPK Signaling Pathway. J Med Food 2024; 27:834-843. [PMID: 38949913 DOI: 10.1089/jmf.2023.k.0292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024] Open
Abstract
Excessive fat deposition leads to obesity and cardiovascular diseases with abnormal metabolism. Pantothenic acid (PA) is a major B vitamin required for energy metabolism. However, the effect of PA on lipid metabolism and obesity has not been explored. We investigated the effects and molecular mechanism of PA on fat accumulation as well as the influence of adipogenic marker genes in both adult male mice and primary adipocytes. First, we demonstrated that PA attenuates weight gain in mice fed high-fat diet (HFD). Besides, PA supplementation substantially improved glucose tolerance and lipid metabolic disorder in obese mice. Furthermore, PA significantly inhibited white adipose tissue (WAT) deposition as well as fat droplets visualized by magnification in both chow and HFD group. More importantly, PA obviously suppressed the mRNA levels of CD36, IL-6, and TNF-α to alleviate inflammation and reduced the levels of PPARγ, aP2, and C/EBPα genes that are related to lipid metabolism in inguinal white adipose tissue (ing-WAT) and epididymal white adipose tissue (ei-WAT). In vitro, PA supplementation showed a lower lipid droplet aggregation as well as reduced expression levels of adipogentic genes. Finally, we identified that PA inhibits the phosphorylation levels of p38 and JNK in murine primary adipocytes. Collectively, our data demonstrated for the first time that PA attenuates lipid metabolic disorder as well as fat deposition by JNK/p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Cunzhen Zhao
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Ziwei Wen
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Yunfei Gao
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Fang Xiao
- Pingqiao District Bureau of Agriculture and Rural Development of Xinyang, Xinyang, China
| | - Jinzhao Yan
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Xiaotong Wang
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Tiantian Meng
- College of Life Science, Xinyang Normal University, Xinyang, China
| |
Collapse
|
47
|
Anaya ES, de Groot EL, Lydon JP, Pangas SA, Hartig SM. Contributions of white adipose tissue to energy requirements for female reproduction. Trends Endocrinol Metab 2024; 35:809-820. [PMID: 38749883 PMCID: PMC11387141 DOI: 10.1016/j.tem.2024.04.012] [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: 02/14/2024] [Revised: 04/13/2024] [Accepted: 04/15/2024] [Indexed: 09/12/2024]
Abstract
Body composition impacts female fertility and there are established relationships between adipose tissue and the reproductive system. Maintaining functional adipose tissue is vital for meeting the energetic demands during the reproductive process, from ovulation to delivery and lactation. White adipose tissue (WAT) shows plastic responses to daily physiology and secretes diverse adipokines that affect the hypothalamic-pituitary-ovarian axis, but many other interorgan interactions remain to be determined. This review summarizes the current state of research on the dialogue between WAT and the female reproductive system, focusing on the impact of this crosstalk on ovarian and endometrial factors essential for fecundity.
Collapse
Affiliation(s)
- Elizabeth S Anaya
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - Evelyn L de Groot
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Stephanie A Pangas
- Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Sean M Hartig
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
48
|
Ahn C, Zhang T, Yang G, Rode T, Varshney P, Ghayur SJ, Chugh OK, Jiang H, Horowitz JF. Years of endurance exercise training remodel abdominal subcutaneous adipose tissue in adults with overweight or obesity. Nat Metab 2024; 6:1819-1836. [PMID: 39256590 DOI: 10.1038/s42255-024-01103-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/09/2024] [Indexed: 09/12/2024]
Abstract
Abnormalities in the structure and metabolic function of abdominal subcutaneous adipose tissue (aSAT) underlie many obesity-related health complications. Endurance exercise improves cardiometabolic health in adults with overweight or obesity, but the effects of endurance training on aSAT are unclear. We included male and female participants who were regular exercisers with overweight or obesity who exercised for >2 years, and cross-sectionally compared them with well-matched non-exercisers with overweight or obesity. Here we show aSAT from exercisers has a higher capillary density, lower Col6a abundance and fewer macrophages compared with non-exercisers. This is accompanied by a greater abundance of angiogenic, ribosomal, mitochondrial and lipogenic proteins. The abundance of phosphoproteins involved in protein translation, lipogenesis and direct regulation of transcripts is also greater in aSAT collected from exercisers. Exploratory ex vivo experiments demonstrate greater angiogenic capacity and higher lipid-storage capacity in samples cultured from aSAT collected from exercisers versus non-exercisers. Regular exercise may play a role in remodelling aSAT structure and proteomic profile in ways that may contribute to preserved cardiometabolic health.
Collapse
Affiliation(s)
- Cheehoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Tao Zhang
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Gayoung Yang
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Thomas Rode
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Pallavi Varshney
- Human Bioenergetics Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Sophia J Ghayur
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Olivia K Chugh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Hui Jiang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
49
|
Jang S, Hong W, Moon Y. Obesity-compromised immunity in post-COVID-19 condition: a critical control point of chronicity. Front Immunol 2024; 15:1433531. [PMID: 39188722 PMCID: PMC11345197 DOI: 10.3389/fimmu.2024.1433531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Post-COVID-19 condition is recognized as a multifactorial disorder, with persistent presence of viral antigens, discordant immunity, delayed viral clearance, and chronic inflammation. Obesity has emerged as an independent risk factor for both SARS-CoV-2 infection and its subsequent sequelae. In this study, we aimed to predict the molecular mechanisms linking obesity and post-COVID-19 distress. Viral antigen-exposed adipose tissues display remarkable levels of viral receptors, facilitating viral entry, deposition, and chronic release of inflammatory mediators and cells in patients. Subsequently, obesity-associated inflammatory insults are predicted to disturb cellular and humoral immunity by triggering abnormal cell differentiation and lymphocyte exhaustion. In particular, the decline in SARS-CoV-2 antibody titers and T-cell exhaustion due to chronic inflammation may account for delayed virus clearance and persistent activation of inflammatory responses. Taken together, obesity-associated defective immunity is a critical control point of intervention against post-COVID-19 progression, particularly in subjects with chronic metabolic distress.
Collapse
Affiliation(s)
- Soonwoo Jang
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
| | - Wooyoung Hong
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
50
|
De Fano M, Malara M, Vermigli C, Murdolo G. Adipose Tissue: A Novel Target of the Incretin Axis? A Paradigm Shift in Obesity-Linked Insulin Resistance. Int J Mol Sci 2024; 25:8650. [PMID: 39201336 PMCID: PMC11354636 DOI: 10.3390/ijms25168650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Adipose tissue (AT) represents a plastic organ that can undergo significant remodeling in response to metabolic demands. With its numerous checkpoints, the incretin system seems to play a significant role in controlling glucose homeostasis and energy balance. The importance of the incretin hormones, namely the glucagon-like peptide-1 (GLP-1) and the glucose-dependent insulinotropic peptide (GIP), in controlling the function of adipose cells has been brought to light by recent studies. Notably, a "paradigm shift" in reevaluating the role of the incretin system in AT as a potential target to treat obesity-linked metabolic disorders resulted from the demonstration that a disruption of the GIP and GLP-1 signaling axis in fat is associated with adiposity-induced insulin-resistance (IR) and/or type 2 diabetes mellitus (T2D). We will briefly discuss the (patho)physiological functions of GLP-1 and GIP signaling in AT in this review, emphasizing their potential impacts on lipid storage, adipogenesis, glucose metabolism and inflammation. We will also address the conundrum with the perturbation of the incretin axis in white or brown fat tissue and the emergence of metabolic disorders. In order to reduce or avoid adiposity-related metabolic complications, we will finally go over a potential scientific rationale for suggesting AT as a novel target for GLP-1 and GIP receptor agonists and co-agonists.
Collapse
Affiliation(s)
- Michelantonio De Fano
- Complex Structure of Endocrinology and Metabolism, Department of Medicine, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, 06081 Perugia, Italy; (M.M.); (C.V.); (G.M.)
| | | | | | | |
Collapse
|