1
|
Seibel AJ, Frosti CL, Tlemçani AR, Lahiri N, Brammer-DePuy JA, Layne MD, Tien J. Obesity-Associated Conditions Hinder Solute Drainage Function of Engineered Human Lymphatic Vessels. Cell Mol Bioeng 2025; 18:53-69. [PMID: 39949491 PMCID: PMC11813835 DOI: 10.1007/s12195-024-00840-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/13/2024] [Indexed: 02/16/2025] Open
Abstract
Purpose Obesity is associated with poor lymphatic solute drainage. It is unclear whether the chronic inflammation, hypoxia, and hyperlipidemia that are together associated with obesity cause impaired drainage function, and if so, whether these conditions act directly on lymphatic endothelial cells (LECs) or are indirectly mediated by the mechanical properties or cellular composition of the surrounding tissue. Methods We engineered blind-ended lymphatic vessels in type I collagen gels and simulated the obese microenvironment with a cocktail of tumor necrosis factor (TNF)-α, cobalt chloride (CoCl2), and oleate, which model inflammation, hypoxia, and hyperlipidemia, respectively. We compared the solute drainage rate and leakage of lymphatics that were exposed to simulated obesity or not. We performed similar assays with lymphatics in stiffened gels, in adipocyte-laden gels, or in the presence of conditioned medium (CM) from adipose cells treated with the same cocktail. Results Lymphatics that were exposed to simulated obesity exhibited more gaps in endothelial junctions, leaked more solute, and drained solute less quickly than control lymphatics did, regardless of matrix stiffness. CM from adipose cells that were exposed to simulated obesity did not affect lymphatics. Lymphatics in adipocyte-laden gels did not exhibit worse drainage function when exposed to simulated obesity. Conclusions The combination of obesity-associated inflammation, hypoxia, and hyperlipidemia impairs lymphatic solute drainage and does so by acting directly on LECs. Surprisingly, adipocytes may play a protective role in preventing obesity-associated conditions from impairing lymphatic solute drainage. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00840-z.
Collapse
Affiliation(s)
- Alex J. Seibel
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215 USA
| | - Cheyanne L. Frosti
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Abderrahman R. Tlemçani
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215 USA
| | - Nikhil Lahiri
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215 USA
| | - Joely A. Brammer-DePuy
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215 USA
| | - Matthew D. Layne
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Joe Tien
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215 USA
- Division of Materials Science and Engineering, Boston University, Boston, MA USA
| |
Collapse
|
2
|
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
|
3
|
Passini FS, Bornstein B, Rubin S, Kuperman Y, Krief S, Masschelein E, Mehlman T, Brandis A, Addadi Y, Shalom SHO, Richter EA, Yardeni T, Tirosh A, De Bock K, Zelzer E. Piezo2 in sensory neurons regulates systemic and adipose tissue metabolism. Cell Metab 2025:S1550-4131(24)00526-6. [PMID: 39919739 DOI: 10.1016/j.cmet.2024.12.016] [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: 04/14/2024] [Revised: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 02/09/2025]
Abstract
Systemic metabolism ensures energy homeostasis through inter-organ crosstalk regulating thermogenic adipose tissue. Unlike the well-described inductive role of the sympathetic system, the inhibitory signal ensuring energy preservation remains poorly understood. Here, we show that, via the mechanosensor Piezo2, sensory neurons regulate morphological and physiological properties of brown and beige fat and prevent systemic hypermetabolism. Targeting runt-related transcription factor 3 (Runx3)/parvalbumin (PV) sensory neurons in independent genetic mouse models resulted in a systemic metabolic phenotype characterized by reduced body fat and increased insulin sensitivity and glucose tolerance. Deletion of Piezo2 in PV sensory neurons reproduced the phenotype, protected against high-fat-diet-induced obesity, and caused adipose tissue browning and beiging, likely driven by elevated norepinephrine levels. Finding that brown and beige fat are innervated by Runx3/PV sensory neurons expressing Piezo2 suggests a model in which mechanical signals, sensed by Piezo2 in sensory neurons, protect energy storage and prevent a systemic hypermetabolic phenotype.
Collapse
Affiliation(s)
- Fabian S Passini
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| | - Bavat Bornstein
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Sarah Rubin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Evi Masschelein
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Tevie Mehlman
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Brandis
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- MICC Cell Observatory, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Huri-Ohev Shalom
- Bert Strassburger Metabolic Center for Preventive Medicine, Sheba Medical Center, Tel-Hashomer, Israel
| | - Erik A Richter
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Tal Yardeni
- Bert Strassburger Metabolic Center for Preventive Medicine, Sheba Medical Center, Tel-Hashomer, Israel
| | - Amir Tirosh
- The Dalia and David Arabov Endocrinology and Diabetes Research Center, Institute of Endocrinology, Sheba Medical Center, Tel-Hashomer, Israel; Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Katrien De Bock
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
4
|
Guo H, Zhang Z, Yang J, Liu J, Lin H, Yin N. Depot-specific acetylation profiles of adipose tissues-therapeutic targets for metabolically unhealthy obesity. Diabetol Metab Syndr 2025; 17:36. [PMID: 39881347 PMCID: PMC11776295 DOI: 10.1186/s13098-025-01599-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/18/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Adipose tissue plays a critical role in the development of metabolically unhealthy obesity (MUO), with distinct adipose depots demonstrating functional differences. This study aimed to investigate the unique characteristics of subcutaneous (SA) and visceral adipose tissue (VA) in MUO. METHODS Paired omental VA and abdominal SA samples were obtained from four male patients with MUO and subjected to Four-Dimensional Data Independent Acquisition (4D-DIA) proteomic and lysine acetylation (Kac) analyses. Differentially expressed proteins and differentially modified Kac sites were identified, quantified, integrated, and subjected to functional analyses. Overlap analysis was performed between our datasets and previously published proteomic datasets in obesity populations. Additionally, differentially modified Kac sites on histones and their related enzymes were identified. RESULTS A total of 281 differentially expressed proteins and 147 differentially modified Kac sites were identified among 6,201 quantifiable proteins and 1,826 quantifiable Kac sites. Upregulated proteins and acetylated proteins in SA were predominantly enriched in extracellular matrix (ECM) remodeling pathways, while those in VA were enriched in energy metabolism and disease-related pathways. Differential ECM remodeling adaptability between SA and VA was primarily mediated by fibranexin and integrin, with COL6A1, COL6A3, and ITGA5 identified as differentially acetylated proteins overlapping between our dataset and previous studies. Potential unique proteins in MUO were enriched in inflammatory processes and closely associated with acetylated modifications. Specific differentially acetylated sites on histones, including H1.2K63, H1XK90, and H3.7K80, showed increased acetylation in VA, with N-deacetylase/N-sulfotransferase 1 (NDST1) identified as the associated enzyme. CONCLUSIONS This study provided a comprehensive dataset on the proteomic and acetylomic profiles of SA and VA, laying a foundation for investigating the pathogenesis and potential therapeutic approaches for MUO. SA was characterized by pronounced ECM remodeling regulation, while VA exhibited poorer adaptability and more prominent metabolic functional changes. These differential processes were influenced not only by protein expression levels but, more importantly, by acetylated modifications. The regulation of acetylated modifications in white adipose tissue (WAT), particularly for the differential Kac sites enriched in ECM remodeling and inflammation-related pathways, may serve as an effective intervention strategy for MUO, with NDST1 emerging as a promising therapeutic target. TRIAL REGISTRATION Not applicable since this study did not involve clinical intervention.
Collapse
Affiliation(s)
- Haoyue Guo
- The Centre for Cleft Lip and Palate Treatment, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Zhiyi Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Juntao Yang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiangfeng Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Hongwei Lin
- The Center for Obesity and Metabolic Disease, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China.
| | - Ningbei Yin
- The Centre for Cleft Lip and Palate Treatment, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China.
| |
Collapse
|
5
|
Ivatt L, Paul M, Miguelez-Crespo A, Hadoke PWF, Bailey MA, Morgan RA, Nixon M. Obesity-induced mesenteric PVAT remodelling is sexually dimorphic, but not driven by ovarian hormones : Short title: Obesity induces sex-specific responses in mesenteric PVAT. Cardiovasc Diabetol 2025; 24:39. [PMID: 39856754 PMCID: PMC11762466 DOI: 10.1186/s12933-025-02596-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Obesity, a major risk factor for cardiovascular disease (CVD), is associated with hypertension and vascular dysfunction. Perivascular adipose tissue (PVAT), a metabolically active tissue surrounding blood vessels, plays a key role in regulating vascular tone. In obesity, PVAT becomes dysregulated which may contribute to vascular dysfunction; how sex impacts the remodelling of PVAT and thus the altered vascular contractility during obesity is unclear. OBJECTIVE To investigate sex-specific PVAT dysregulation in the setting of obesity as a potential driver of sex differences in vascular pathologies and CVD risk. METHODS Adult male and female C57Bl/6J mice were fed an obesogenic high-fat diet (HFD) or regular chow for 16 weeks. Mesenteric PVAT (mPVAT) was isolated for RNA-sequencing and histological analysis, and mesenteric arteries were isolated for assessment of vascular function by wire myography. In a separate study, female mice were subjected to bilateral ovariectomy prior to dietary intervention to determine the contribution of ovarian hormones to PVAT dysregulation. RESULTS Transcriptomic analysis of mPVAT revealed sexually dimorphic responses to HFD, with upregulation of extracellular matrix (ECM) remodelling pathways in male but not female mice. Histological and RT-qPCR approaches demonstrated increased collagen deposition and ECM remodelling in mPVAT from obese male compared with obese female mice. Assessment of vascular function in mesenteric arteries -/+ PVAT revealed that in obesity, mPVAT impaired endothelium-mediated vasodilation in male but not female mice. Ovariectomy of female mice prior to HFD administration did not alter ECM transcript expression or collagen deposition in mPVAT compared to sham-operated female mice. CONCLUSIONS Obesity induces sex-specific molecular remodelling in mPVAT, with male mice exhibiting unique upregulation of ECM pathways and increased collagen deposition compared to females. Moreover, the relative protection of female mice from obesity-induced mPVAT dysregulation is not mediated by ovarian hormones. These data highlight a potential sex-specific mechanistic link between mPVAT and mesenteric artery dysfunction in obesity, and provides crucial insights for future development of treatment strategies that consider the unique cardiovascular risks in men and women.
Collapse
Affiliation(s)
- Lisa Ivatt
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Mhairi Paul
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | | | - Patrick W F Hadoke
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Matthew A Bailey
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | | | - Mark Nixon
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK.
| |
Collapse
|
6
|
Lee Y, Tukei KL, Fang Y, Kuila S, Liu X, Imoukhuede PI. Integrative analysis of angiogenic signaling in obesity: capillary features and VEGF binding kinetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.23.630107. [PMID: 39763822 PMCID: PMC11703262 DOI: 10.1101/2024.12.23.630107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Obesity is a global health crisis, with its prevalence particularly severe in the United States, where over 42% of adults are classified as obese. Obesity is driven by complex molecular and tissue-level mechanisms that remain poorly understood. Among these, angiogenesis-primarily mediated by vascular endothelial growth factor (VEGF-A)-is critical for adipose tissue expansion but presents unique challenges for therapeutic targeting due to its intricate regulation. Systems biology approaches have advanced our understanding of VEGF-A signaling in vascular diseases, but their application to obesity is limited by scattered and sometimes contradictory data. To address this gap, we performed a comprehensive analysis of the existing literature to synthesize key findings, standardize data, and provide a holistic perspective on the adipose vascular microenvironment. The data mining revealed five key findings: (1) obesity increases adipocyte size by 78%; (2) vessel density in adipose tissue decreases by 51% in obese mice, with vessels being 47-58% smaller and 4-9 times denser in comparison with tumor vessels; (3) capillary basement membrane thickness remains similar regardless of obesity; (4) VEGF-A shows the strongest binding affinity for VEGFR1, with four times stronger affinity for VEGFR2 than for NRP1; and (5) binding affinities measured by radioligand binding assay and surface plasmon resonance (SPR) are significantly different. These consolidated findings provide essential parameters for systems biology modeling, new insights into obesity-induced changes in adipose tissue, and a foundation for developing angiogenesis-targeting therapies for obesity.
Collapse
|
7
|
Schleh MW, Ryan BJ, Ahn C, Ludzki AC, Van Pelt DW, Pitchford LM, Chugh OK, Luker AT, Luker KE, Samovski D, Abumrad NA, Burant CF, Horowitz JF. Impaired suppression of fatty acid release by insulin is a strong predictor of reduced whole-body insulin-mediated glucose uptake and skeletal muscle insulin receptor activation. Acta Physiol (Oxf) 2025; 241:e14249. [PMID: 39487600 DOI: 10.1111/apha.14249] [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: 06/26/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 11/04/2024]
Abstract
AIM To examine factors underlying why most, but not all, adults with obesity exhibit impaired insulin-mediated glucose uptake, we compared: (1) adipose tissue fatty acid (FA) release, (2) skeletal muscle lipid droplet (LD) characteristics, and (3) insulin signalling events, in skeletal muscle of adults with obesity with relatively high versus low insulin-mediated glucose uptake. METHODS Seventeen adults with obesity (BMI: 36 ± 3 kg/m2) completed a 2 h hyperinsulinemic-euglycemic clamp with stable isotope tracer infusions to measure glucose rate of disappearance (glucose Rd) and FA rate of appearance (FA Ra). Skeletal muscle biopsies were collected at baseline and 30 min into the insulin infusion. Participants were stratified into HIGH (n = 7) and LOW (n = 10) insulin sensitivity cohorts by their glucose Rd during the hyperinsulinemic clamp (LOW< 400; HIGH >550 nmol/kgFFM/min/[μU/mL]). RESULTS Insulin-mediated suppression of FA Ra was lower in LOW compared with HIGH (p < 0.01). In skeletal muscle, total intramyocellular lipid content did not differ between cohorts. However, the size of LDs in the subsarcolemmal region (SS) of type II muscle fibres was larger in LOW compared with HIGH (p = 0.01). Additionally, insulin receptor-β (IRβ) interactions with regulatory proteins CD36 and Fyn were lower in LOW versus HIGH (p < 0.01), which aligned with attenuated insulin-mediated Tyr phosphorylation of IRβ and downstream insulin-signalling proteins in LOW. CONCLUSION Collectively, reduced ability for insulin to suppress FA mobilization, with accompanying modifications in intramyocellular LD size and distribution, and diminished IRβ interaction with key regulatory proteins may be key contributors to impaired insulin-mediated glucose uptake commonly found in adults with obesity.
Collapse
Affiliation(s)
- Michael W Schleh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Benjamin J Ryan
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Cheehoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alison C Ludzki
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Douglas W Van Pelt
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lisa M Pitchford
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Olivia K Chugh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Austin T Luker
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Dmitri Samovski
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nada A Abumrad
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Charles F Burant
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
8
|
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
|
9
|
Fallone F, Rebeaud M, Bouche C, Fontaine J, Arellano C, Ducoux-Petit M, Orgerit L, Deudon R, Nicolle R, Franchet C, Estève D, Mouton-Barbosa E, Dauvillier S, Moutahir M, Burlet-Schiltz O, Bouloumié A, Vaysse C, Muller C. Lack of fibro-inflammatory response in human mammary adipose tissue in obesity. Int J Obes (Lond) 2024:10.1038/s41366-024-01705-1. [PMID: 39738492 DOI: 10.1038/s41366-024-01705-1] [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: 04/29/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Understanding how obesity impacts human mammary adipose tissue (MAT) biology is crucial for deciphering its role in mammary epithelium during both physiological and pathophysiological processes, including breast cancer. Hypertrophic mammary adipocytes and Crown-Like Structures are present in MAT of patients with obesity but whether these changes initiate a fibro-inflammatory response at the tissue level remains insufficiently explored. OBJECTIVE We investigated the markers of adipose tissue dysfunction (immune cell infiltration, secretion pattern and fibrosis) in tumor-free MAT of patients with obesity versus patients who are lean. METHODS Tumor-free MAT were obtained from 96 women with (n = 43) or without (n = 53) obesity who underwent mastectomy for breast cancer risk reduction or treatment. Immune and non-immune cell infiltration were determined using flow cytometry. Bulk transcriptomic was used to characterize the phenotype of CD206+ macrophages whose infiltration is increased in patients with obesity. Conditioned-medium were prepared from MAT to characterize their secretome and dose adipokines and cytokines by ELISA assay. The extra-cellular matrix (ECM) deposition was evaluated by Masson trichrome staining on cross-stained sections, 3D imaging of red picrosirius-stained tissues and measure of hydroxyproline content. RESULTS We observed an increase of CD206+/HLA-DR+ macrophages in the stromal vascular fraction of MAT from patients with obesity compared to patients who are lean. Other immune cell infiltration and endothelial or adipose progenitor cell numbers were similar between groups. Bulk transcriptomics on CD206+ macrophages revealed a significant decrease in ECM component expression and processing in obesity. In addition, no heightened secretion of pro-inflammatory cytokines, TGF-β1 or MCP-1 was observed in the samples from patients with obesity. ECM characterization revealed an absence of fibrosis, with MAT of patients with obesity showing even a slightly reduced collagen secretion and deposition compared with their lean counterparts. CONCLUSIONS Obesity is not associated with inflammation nor fibrosis in MAT, highlighting its unique behavior.
Collapse
Affiliation(s)
- Frédérique Fallone
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France.
| | - Marie Rebeaud
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Caroline Bouche
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Jessica Fontaine
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université de Toulouse, INSERM, UPS, Toulouse, France
| | - Carlo Arellano
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Manuelle Ducoux-Petit
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Infrastructure Nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Lucyle Orgerit
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Rémi Deudon
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Rémy Nicolle
- Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018, Paris, France
| | - Camille Franchet
- Département d'Anatomo-Pathologie, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - David Estève
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Emmanuelle Mouton-Barbosa
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Infrastructure Nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Stéphanie Dauvillier
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Mohamed Moutahir
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Infrastructure Nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Anne Bouloumié
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université de Toulouse, INSERM, UPS, Toulouse, France
| | - Charlotte Vaysse
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Catherine Muller
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France.
| |
Collapse
|
10
|
Do A, Zahrawi F, Mehal WZ. Therapeutic landscape of metabolic dysfunction-associated steatohepatitis (MASH). Nat Rev Drug Discov 2024:10.1038/s41573-024-01084-2. [PMID: 39609545 DOI: 10.1038/s41573-024-01084-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/30/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and its severe subgroup metabolic dysfunction-associated steatohepatitis (MASH) have become a global epidemic and are driven by chronic overnutrition and multiple genetic susceptibility factors. The physiological outcomes include hepatocyte death, liver inflammation and cirrhosis. The first therapeutic for MASLD and MASH, resmetirom, has recently been approved for clinical use and has energized this therapeutic space. However, there is still much to learn in clinical studies of MASH, such as the scale of placebo responses, optimal trial end points, the time required for fibrosis reversal and side effect profiles. This Review introduces aspects of disease pathogenesis related to drug development and discusses two main therapeutic approaches. Thyroid hormone receptor-β agonists, such as resmetirom, as well as fatty acid synthase inhibitors, target the liver and enable it to function within a toxic metabolic environment. In parallel, incretin analogues such as semaglutide improve metabolism, allowing the liver to self-regulate and reversing many aspects of MASH. We also discuss how combinations of therapeutics could potentially be used to treat patients.
Collapse
Affiliation(s)
- Albert Do
- Section of Digestive Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Division of Gastroenterology, University of California, Davis, Davis, USA
| | - Frhaan Zahrawi
- Section of Digestive Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Wajahat Z Mehal
- Section of Digestive Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- West Haven Veterans Hospital, West Haven, CT, USA.
| |
Collapse
|
11
|
Aboy-Pardal MCM, Guadamillas MC, Guerrero CR, Català-Montoro M, Toledano-Donado M, Terrés-Domínguez S, Pavón DM, Jiménez-Jiménez V, Jimenez-Carretero D, Zamai M, Folgueira C, Cerezo A, Lolo FN, Nogueiras R, Sabio G, Sánchez-Álvarez M, Echarri A, Garcia R, Del Pozo MA. Plasma membrane remodeling determines adipocyte expansion and mechanical adaptability. Nat Commun 2024; 15:10102. [PMID: 39609408 PMCID: PMC11605069 DOI: 10.1038/s41467-024-54224-y] [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: 06/02/2022] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
Adipocytes expand massively to accommodate excess energy stores and protect the organism from lipotoxicity. Adipose tissue expandability is at the center of disorders such as obesity and lipodystrophy; however, little is known about the relevance of adipocyte biomechanics on the etiology of these conditions. Here, we show in male mice in vivo that the adipocyte plasma membrane undergoes caveolar domain reorganization upon lipid droplet expansion. As the lipid droplet grows, caveolae disassemble to release their membrane reservoir and increase cell surface area, and transfer specific caveolar components to the LD surface. Adipose tissue null for caveolae is stiffer, shows compromised deformability, and is prone to rupture under mechanical compression. Mechanistically, phosphoacceptor Cav1 Tyr14 is required for caveolae disassembly: adipocytes bearing a Tyr14Phe mutation at this residue are stiffer and smaller, leading to decreased adiposity in vivo; exhibit deficient transfer of Cav1 and EHD2 to the LD surface, and show distinct Cav1 molecular dynamics and tension adaptation. These results indicate that Cav1 phosphoregulation modulates caveolar dynamics as a relevant component of the homeostatic mechanoadaptation of the differentiated adipocyte.
Collapse
Affiliation(s)
- María C M Aboy-Pardal
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marta C Guadamillas
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Science and Agroforestal Technology and Genetics, Faculty of Biochemistry and Environmental Sciences, University of Castilla-La Mancha, Toledo, Spain
| | - Carlos R Guerrero
- ForceTool group, Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, Madrid, Spain
| | - Mauro Català-Montoro
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Mónica Toledano-Donado
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sara Terrés-Domínguez
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Dácil M Pavón
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Allergy Therapeutics, Avenida Punto Es, 12, 28805 Alcalá de Henares, Madrid, Spain
| | - Víctor Jiménez-Jiménez
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Health Science, Universidad Católica Santa Teresa de Jesús de Ávila, Ávila, Spain
| | - Daniel Jimenez-Carretero
- Cellomics Unit, Cell and Developmental Biology Area. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Moreno Zamai
- Microscopy and Dynamic Imaging Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Cintia Folgueira
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas CIMUS, Santiago de Compostela, Spain
- Stress kinases in Diabetes, Cancer and Cardiovascular Disease lab. Cardiovascular risk factors & brain function program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ana Cerezo
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Molecular Pharmacology, Lilly Research Laboratories, Alcobendas, Spain
| | - Fidel-Nicolás Lolo
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Rubén Nogueiras
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas CIMUS, Santiago de Compostela, Spain
| | - Guadalupe Sabio
- Stress kinases in Diabetes, Cancer and Cardiovascular Disease lab. Cardiovascular risk factors & brain function program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Cell Compartmentalization, Homeostasis and Inflammation lab, Department of Metabolic and Inflammatory Diseases. Instituto de Investigaciones Biomédicas "Sols-Morreale"-CSIC, Madrid, Spain
| | - Asier Echarri
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Mechanobiology of Organelles lab. Department of Cellular and Molecular Biology. Centro de Investigaciones Biológicas Margarita Salas - CSIC, Madrid, Spain
| | - Ricardo Garcia
- ForceTool group, Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, Madrid, Spain
| | - Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
12
|
Wang Y, Zhang Y, Leung V, Heydari Seradj S, Sonmez U, Servin-Vences R, Lipomi D, Ye L, Patapoutian A. A key role of PIEZO2 mechanosensitive ion channel in adipose sensory innervation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.624210. [PMID: 39605632 PMCID: PMC11601537 DOI: 10.1101/2024.11.18.624210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Compared to the well-established functions of sympathetic innervation, the role of sensory afferents in adipose tissues remains less understood. Recent work revealed the anatomical and physiological significance of adipose sensory innervation; however, its molecular underpinning remains unclear. Here, using organ-targeted single-cell RNA sequencing, we identified the mechanoreceptor PIEZO2 as one of the most prevalent receptors in fat-innervating dorsal root ganglia (DRG) neurons. We found that selective PIEZO2 deletion in fat-innervating neurons phenocopied the molecular alternations in adipose tissue caused by DRG ablation. Conversely, a gain-of-function PIEZO2 mutant shifted the adipose phenotypes in the opposite direction. These results indicate that PIEZO2 plays a major role in the sensory regulation of adipose tissues. This discovery opens new avenues for exploring mechanosensation in organs not traditionally considered mechanically active, such as the adipose tissues, and therefore sheds light on the broader significance of mechanosensation in regulating organ function and homeostasis.
Collapse
|
13
|
Park S, Kim Y, Nam D, Lee J, Song SH. Superficial venous morphometry in the antecubital fossa: An autonomous robotic ultrasound-based analysis. J Vasc Access 2024; 25:1793-1800. [PMID: 37448206 DOI: 10.1177/11297298231186651] [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: 07/15/2023] Open
Abstract
BACKGROUND The antecubital fossa is an important site for venepuncture and intravenous procedures. The size and location of a vein can affect the success of venepuncture and intravenous access. Several studies have investigated the superficial vein morphometry, but they had small sample sizes or focused on specific populations or groups. Therefore, we conducted a prospective study with large participants in general population to analyse the morphology of the antecubital superficial vein and identify the association of sex, age and body mass index (BMI) with the size and location of the vein. METHODS This study collected images of superficial veins prospectively using autonomous robotic ultrasound on the antecubital area between October and November 2020. We measured the superficial vein depth, vertical diameter and horizontal diameter at the antecubital area, extracted population characteristics (sex, age and BMI), and analysed a relationship between the vein dimensions and the characteristics. RESULTS In this study, data from 461 participants (201 males and 260 females) with mean age of 41.1 years were produced. The mean vein depth, mean vertical diameter and mean horizontal diameter (±standard deviation) were 4.81 (±2.17), 3.01 (±1.10) and 4.46 (±1.60) mm, respectively. We found significant differences in vein dimensions between males and females, with males having larger vertical and horizontal diameters than females (p < 0.001). The study also revealed significant differences in vein depth and dimensions among age groups and BMI subgroups (p < 0.001). CONCLUSIONS These findings revealed that the superficial vein in the antecubital area was oval, with a larger horizontal diameter than vertical diameter. Morphometry revealed differences in sex, age and BMI. Understanding variations in vein dimensions among different subgroups can help medical professionals improve success rate of venous access and patient safety.
Collapse
Affiliation(s)
| | | | | | - Jooran Lee
- Division of Life Science and Department of Chemical and Biological Engineering, The Hong Kong University of Science of Technology, Hong Kong SAR, China
| | - Sang Hoon Song
- Department of Laboratory Medicine, Seoul National University Hospital and College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
14
|
Nah J, Yun N, Yoo H, Park S, Pae M. Time-Restricted Feeding Attenuates Adipose Tissue Inflammation and Fibrosis in Mice Under Chronic Light Exposure. Int J Mol Sci 2024; 25:11524. [PMID: 39519077 PMCID: PMC11546375 DOI: 10.3390/ijms252111524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Time-restricted feeding (TRF) has emerged as a promising dietary approach for improving metabolic parameters associated with obesity. However, it remains largely unclear whether TRF offers benefits for obesity related to exposure to light at night. This study examined whether lean and obese mice under chronic light exposure could benefit from TRF intervention. Six-week-old C57BL/6 male mice were fed either a low-fat diet or a high-fat diet under a 12 h light/12 h dark cycle for 6 weeks. They were then divided into three subgroups: control light, chronic 24 h light, and chronic light with a daily 10 h TRF. Chronic light exposure led to increased weight gain and higher expression of inflammatory and fibrotic markers in the adipose tissue of both lean and obese mice. It also increased hepatic triglyceride content in mice, regardless of their weight status. TRF protected both lean and obese mice from weight gain, normalized inflammatory and fibrotic gene expression, and reduced adipose tissue collagen and liver triglyceride accumulation caused by light exposure alone or in combination with obesity. These results suggest that TRF could have clinical implications for preventing obesity associated with night shift work, regardless of current weight status.
Collapse
Affiliation(s)
| | | | | | | | - Munkyong Pae
- Department of Food and Nutrition, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju 28644, Republic of Korea; (J.N.); (N.Y.); (H.Y.); (S.P.)
| |
Collapse
|
15
|
Contessi Negrini N, Pellegrinelli V, Salem V, Celiz A, Vidal-Puig A. Breaking barriers in obesity research: 3D models of dysfunctional adipose tissue. Trends Biotechnol 2024:S0167-7799(24)00278-6. [PMID: 39443224 DOI: 10.1016/j.tibtech.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Obesity is a global health crisis characterised by excessive accumulation of adipose tissue (AT). Under obesogenic conditions, this metabolically active tissue undergoes fibrosis and inflammation, leading to obesity-linked comorbidities. Modelling AT is essential for understanding its pathophysiology and developing treatments to protect against metabolic complications. 3D in vitro AT models are promising tools that address the limitations of traditional 2D in vitro models and in vivo animal models, providing enhanced biomimetic and human-relevant platforms. 3D models facilitate the study of AT pathophysiology and therapeutic screening. This review discusses the crucial role of AT in obesity-linked comorbidities, its dynamicity and complexity, and recent advances in engineering 3D scaffold-based in vitro dysfunctional AT models, highlighting potential breakthroughs in metabolic research and beyond.
Collapse
Affiliation(s)
- Nicola Contessi Negrini
- Department of Bioengineering, Imperial College London, London, UK; The Francis Crick Institute, London, UK.
| | | | - Victoria Salem
- Department of Bioengineering, Imperial College London, London, UK
| | - Adam Celiz
- Department of Bioengineering, Imperial College London, London, UK; The Francis Crick Institute, London, UK
| | - Antonio Vidal-Puig
- MRC Institute of Metabolic Science and Medical Research Council, Cambridge, UK; Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, PR China; Centro de Investigacion Principe Felipe (CIPF), Valencia, Spain; Cambridge Heart and Lung Research Institute, Cambridge, UK
| |
Collapse
|
16
|
Baudin J, Hernandez-Baixauli J, Romero-Giménez J, Yang H, Mulero F, Puiggròs F, Mardinoglu A, Arola L, Caimari A. A cocktail of histidine, carnosine, cysteine and serine reduces adiposity and improves metabolic health and adipose tissue immunometabolic function in ovariectomized rats. Biomed Pharmacother 2024; 179:117326. [PMID: 39208671 DOI: 10.1016/j.biopha.2024.117326] [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: 05/16/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Many women have sought alternative therapies to address menopause. Recently, a multi-ingredient supplement (MIS) containing L-histidine, L-carnosine, L-serine, and L-cysteine has been shown to be effective at ameliorating hepatic steatosis (HS) in ovariectomized (OVX) rats, a postmenopausal oestrogen deficiency model. Considering that HS frequently accompanies obesity, which often occurs during menopause, we aimed to investigate the effects of this MIS for 8 weeks in OVX rats. Twenty OVX rats were orally supplemented with either MIS (OVX-MIS) or vehicle (OVX). Ten OVX rats received vehicle orally along with subcutaneous injections of 17β-oestradiol (OVX-E2), whereas 10 rats underwent a sham operation and received oral and injected vehicles (control group). MIS consumption partly counteracted the fat mass accretion observed in OVX animals, leading to decreased total fat mass, adiposity index and retroperitoneal white adipose tissue (RWAT) adipocyte hypertrophy. OVX-MIS rats also displayed increased lean mass and lean/fat ratio, suggesting a healthier body composition, similar to the results reported for OVX-E2 animals. MIS consumption decreased the circulating levels of the proinflammatory marker CRP, the total cholesterol-to-HDL-cholesterol ratio and the leptin-to-adiponectin ratio, a biomarker of diabetes risk and metabolic syndrome. RWAT transcriptomics indicated that MIS favourably regulated genes involved in adipocyte structure and morphology, cell fate determination and differentiation, glucose/insulin homeostasis, inflammation, response to stress and oxidative phosphorylation, which may be mechanisms underlying the beneficial effects described for OVX-MIS rats. Our results pave the way for using this MIS formulation to improve the body composition and immunometabolic health of menopausal women.
Collapse
Affiliation(s)
- Julio Baudin
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, Reus 43204, Spain; Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona 43007, Spain
| | - Julia Hernandez-Baixauli
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, Reus 43204, Spain
| | - Jordi Romero-Giménez
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, Reus 43204, Spain
| | - Hong Yang
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm SE-17165, Sweden
| | - Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Francesc Puiggròs
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, Reus 43204, Spain
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm SE-17165, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom
| | - Lluís Arola
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona 43007, Spain.
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, Reus 43204, Spain.
| |
Collapse
|
17
|
Ke S, Hu Q, Zhu G, Li L, Sun X, Cheng H, Li L, Yao Y, Li H. Remodeling of white adipose tissue microenvironment against obesity by phytochemicals. Phytother Res 2024; 38:4904-4922. [PMID: 36786412 DOI: 10.1002/ptr.7758] [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: 08/31/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 02/15/2023]
Abstract
Obesity is a kind of chronic disease due to a long-term imbalance between energy intake and expenditure. In recent years, the number of obese people around the world has soared, and obesity problem should not be underestimated. Obesity is characterized by changes in the adipose microenvironment, mainly manifested as hypertrophy, chronic inflammatory status, hypoxia, and fibrosis, thus contributing to the pathological changes of other tissues. A plethora of phytochemicals have been found to improve adipose microenvironment, thus prevent and resist obesity, providing a new research direction for the treatment of obesity and related diseases. This paper discusses remodeling of the adipose tissue microenvironment as a therapeutic avenue and reviews the progress of phytochemicals in fighting obesity by improving the adipose microenvironment.
Collapse
Affiliation(s)
- Shuwei Ke
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, People's Republic of China
| | - Qingyuan Hu
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, People's Republic of China
| | - Guanyao Zhu
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, People's Republic of China
| | - Linghuan Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, People's Republic of China
| | - Xuechao Sun
- Research and Development Department, Zhejiang Starry Pharmaceutical Co., Ltd., Taizhou, People's Republic of China
| | - Hongbin Cheng
- Research and Development Department, Zhejiang Starry Pharmaceutical Co., Ltd., Taizhou, People's Republic of China
| | - Lingqiao Li
- Research and Development Department, Zhejiang Starry Pharmaceutical Co., Ltd., Taizhou, People's Republic of China
| | - Yuanfa Yao
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, People's Republic of China
| | - Hanbing Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, People's Republic of China
| |
Collapse
|
18
|
Lee S, Cho YK, Kim H, Choi C, Kim S, Lee YH. miR-10a regulates cell death and inflammation in adipose tissue of male mice with diet-induced obesity. Mol Metab 2024; 90:102039. [PMID: 39342992 PMCID: PMC11513492 DOI: 10.1016/j.molmet.2024.102039] [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: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024] Open
Abstract
OBJECTIVE Adipose tissue remodeling plays a critical role in obesity-induced metabolic dysfunction, but the underlying molecular mechanisms remain incompletely understood. This study investigates the role of miR-10a-5p in adipose tissue inflammation and metabolic dysfunction induced by a high-fat diet (HFD). METHODS Male miR-10a knockout (KO) mice were fed a HFD to induce obesity for up to 16 weeks. RNA sequencing (RNA-seq) analysis was performed to profile mRNA expression and assess the effects of miR-10a-5p KO in gonadal white adipose tissue (gWAT). Additional analyses included immunoblotting, qPCR, histological examination, and validation of the miR-10a-5p target sequence using a dual-luciferase reporter assay. RESULTS miR-10a-5p was highly expressed in gWAT but decreased after 8 weeks of HFD feeding. Over the 16-week HFD period, miR-10a KO mice exhibited greater weight gain and reduced energy expenditure compared to wild-type (WT) controls. gWAT of miR-10a KO mice on a HFD showed an increased population of proinflammatory macrophages, elevated inflammation, and increased cell death, characterized by upregulated apoptosis and necrosis markers. This was also associated with increased triglyceride accumulation in liver. Mechanistically, the proapoptotic gene Bcl2l11 was identified as a direct target of miR-10a-5p. Loss of miR-10a-5p led to BIM-mediated adipocyte death and inflammation, contributing to mitochondrial metabolic dysregulation, increased fibrosis marker expression, and the onset of inflammation in adipose tissue. CONCLUSIONS This study demonstrates the significant role of miR-10a-5p and its downstream target BIM in regulating adipocyte death during diet-induced obesity. This signaling pathway presents a potential therapeutic target for modulating obesity-induced inflammation and cell death in adipose tissue.
Collapse
Affiliation(s)
- Sumin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoon Keun Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Heeseong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Cheoljun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sangseob Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yun-Hee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
19
|
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
|
20
|
Moqaddam MA, Nemati M, Dara MM, Hoteit M, Sadek Z, Ramezani A, Rand MK, Abbassi-Daloii A, Pashaei Z, Almaqhawi A, Razi O, Escobar KA, Supriya R, Saeidi A, Zouhal H. Exploring the Impact of Astaxanthin Supplementation in Conjunction with a 12-Week CrossFit Training Regimen on Selected Adipo-Myokines Levels in Obese Males. Nutrients 2024; 16:2857. [PMID: 39275173 PMCID: PMC11397083 DOI: 10.3390/nu16172857] [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: 06/11/2024] [Revised: 07/22/2024] [Accepted: 08/14/2024] [Indexed: 09/16/2024] Open
Abstract
OBJECTIVE Obesity is associated with an exacerbated metabolic condition that is mediated through impairing balance in the secretion of some adipo-myokines. Therefore, the objective of the present study was to explore the impact of astaxanthin supplementation in conjunction with a 12-week CrossFit training regimen on some selected adipo-myokines, insulin insensitivity, and serum lipid levels in obese males. MATERIAL AND METHODS This study is a randomized control trial design; 60 obese males were randomly divided into four groups of 15, including the control group (CG), supplement group (SG), training group (TG), and combined training and supplement group (TSG). The participants were subjected to 12 weeks of astaxanthin (AST) supplementation [20 mg/d capsule, once/d] or CrossFit training or a combination of both interventions. The training regimen comprised 36 sessions of CrossFit, each lasting 60 min, conducted three times per week. The metabolic indices, body composition, anthropometrical, cardio-respiratory, and also some plasma adipo-myokine factors, including decorin (DCN), activin A, myostatin (MST), transforming growth factor (TGF)-β1, and follistatin (FST), were examined 12 and 72 h before the initiation of the main interventional protocols, and then 72 h after the final session of the training protocol. RESULTS There was no significant difference in the baseline data between the groups (p > 0.05). There were significant interactions between group x time for DCN (η2 = 0.82), activin A (η2 = 0.50), FST (η2 = 0.92), MST (η2 = 0.75), and TGFB-1 (η2 = 0.67) (p < 0.001 for all the variables). Significantly changes showed for DCN in TSG compared to TG and SG and also TG compared to SG (p = 0.0001); for activin A in SG compared to TG (p = 0.01) and TSG (p = 0.002); for FST in SG compared to TG and TSG (p = 0.0001), also in TSG compared to TG (p = 0.0001); for MST in SG, TG, and TSG compared to CG (p = 0.0001) and also in TSG compared to SG (p = 0.0001) and TG (p = 0.001); for TGFB-1 in SG, TG, and TSG compared to CG (p = 0.0001) and also TSG compared to SG (p = 0.0001) and TG (p = 0.001). CONCLUSIONS The 12-week CrossFit training concurrent with AST supplementation reduced anthropometric and metabolic factors and also serum lipid levels while producing positive changes in body composition and cardiovascular factors. Increased FST and DCN and reduced activin A, MST, and TGF-β1 were other affirmative responses to both interventions.
Collapse
Affiliation(s)
- Mohammad Ahmadi Moqaddam
- Department of Physical Education and Sport Science, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Morteza Nemati
- Department of Biomechanics and Sports Injuries, Faculty of Physical Education and Sports Sciences, Kharazmi University, Tehran 1571914911, Iran
| | - Marjan Mansouri Dara
- Department of Physical Education and Sport Science, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Maha Hoteit
- Food Science Unit, National Council for Scientific Research of Lebanon (CNRS-L), Beirut 11-8281, Lebanon
- Section 1, Faculty of Public Health, Lebanese University, Beirut 6573, Lebanon
| | - Zahra Sadek
- Section 1, Faculty of Public Health, Lebanese University, Beirut 6573, Lebanon
- Laboratory of Motor System, Handicap and Rehabilitation (MOHAR), Faculty of Public Health, Lebanese University, Beirut 6573, Lebanon
| | - Akbar Ramezani
- Ayatollah Amoli Branch, Department of Exercise Physiology, Islamic Azad University, Amol 6134937333, Iran
| | - Mahboubeh Khak Rand
- Ayatollah Amoli Branch, Department of Exercise Physiology, Islamic Azad University, Amol 6134937333, Iran
| | - Asieh Abbassi-Daloii
- Ayatollah Amoli Branch, Department of Exercise Physiology, Islamic Azad University, Amol 6134937333, Iran
| | - Zhaleh Pashaei
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, University of Tabriz, Tabriz 5166616471, Iran
| | - Abdullah Almaqhawi
- Department of Family Medicine and Community, College of Medicine, King Faisal University, Al Ahsa 31982, Saudi Arabia
| | - Omid Razi
- Department of Exercise Physiology, Faculty of Physical Education and Sports Science, Razi University, Kermanshah 6714414971, Iran
| | - Kurt A Escobar
- Department of Kinesiology, California State University, Long Beach, CA 90840, USA
| | - Rashmi Supriya
- Centre for Health and Exercise Science Research, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
- Academy of Wellness and Human Development, Faculty of Arts and Social Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
| | - Ayoub Saeidi
- Department of Physical Education and Sport Sciences, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj 1517566177, Iran
| | - Hassane Zouhal
- M2S (Laboratoire Mouvement, Sport, Santé)-EA 1274, Université Rennes, 35044 Rennes, France
- Institut International des Sciences du Sport (2I2S), 35850 Irodouer, France
| |
Collapse
|
21
|
Luca T, Pezzino S, Puleo S, Castorina S. Lesson on obesity and anatomy of adipose tissue: new models of study in the era of clinical and translational research. J Transl Med 2024; 22:764. [PMID: 39143643 PMCID: PMC11323604 DOI: 10.1186/s12967-024-05547-3] [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: 03/10/2024] [Accepted: 07/28/2024] [Indexed: 08/16/2024] Open
Abstract
Obesity is a serious global illness that is frequently associated with metabolic syndrome. Adipocytes are the typical cells of adipose organ, which is composed of at least two different tissues, white and brown adipose tissue. They functionally cooperate, interconverting each other under physiological conditions, but differ in their anatomy, physiology, and endocrine functions. Different cellular models have been proposed to study adipose tissue in vitro. They are also useful for elucidating the mechanisms that are responsible for a pathological condition, such as obesity, and for testing therapeutic strategies. Each cell model has its own characteristics, culture conditions, advantages and disadvantages. The choice of one model rather than another depends on the specific study the researcher is conducting. In recent decades, three-dimensional cultures, such as adipose spheroids, have become very attractive because they more closely resemble the phenotype of freshly isolated cells. The use of such models has developed in parallel with the evolution of translational research, an interdisciplinary branch of the biomedical field, which aims to learn a scientific translational approach to improve human health and longevity. The focus of the present review is on the growing body of data linking the use of new cell models and the spread of translational research. Also, we discuss the possibility, for the future, to employ new three-dimensional adipose tissue cell models to promote the transition from benchside to bedsite and vice versa, allowing translational research to become routine, with the final goal of obtaining clinical benefits in the prevention and treatment of obesity and related disorders.
Collapse
Affiliation(s)
- Tonia Luca
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy.
| | | | - Stefano Puleo
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| |
Collapse
|
22
|
Zelicha H, Bell DS, Chen D, Chen Y, Livingston EH. Obesity and abdominal hernia in ambulatory patients, 2018-2023. Hernia 2024; 28:1317-1324. [PMID: 38795218 PMCID: PMC11297064 DOI: 10.1007/s10029-024-03034-8] [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: 01/11/2024] [Accepted: 03/28/2024] [Indexed: 05/27/2024]
Abstract
PURPOSE To determine the relationship between abdominal hernia and obesity. Although obesity is frequently cited as a risk factor for abdominal hernia, few studies have confirmed this association (Menzo et al. Surg Obes Relat Dis 14:1221-1232. 10.1016/j.soard.2018.07.005, 2018). METHODS A cross-sectional study of primary care ambulatory patients aged older than 16 years treated at UCLA Health from 01/01/2018 to 06/06/2023. Abdominal hernia was identified by clinic encounter ICD-10 codes (K40-K46). RESULTS There were 41,703 hernias identified among 1,362,440 patients (306.1 per10,000) with a mean age of 62.5 ± 16.1 years, and 57.6% were men. Nearly half (44.7%) of all abdominal hernias were diaphragmatic. There was an approximately equal distribution of the ventral (28.7%) and inguinal (24.3%) hernia. Each hernia type had a different relationship with obesity: The odds of having a ventral hernia increased with BMI in both sexes: BMI 25-29.9 kg/m2 odds ratio (OR) = 1.65, (CI 1.56-1.74); BMI 30-39.9 kg/m2 OR = 2.42 (CI 2.29-2.56), BMI 40-49.9 kg/m2 OR = 2.28 (CI 2.05-2.54) and BMI > = 50 kg/m2 OR = 2.54 (CI 2.03-3.17) all relative to normal BMI. In contrast, the odds of having an inguinal hernia decreased with obesity relative to normal weight [obesity (BMI 30-39.9 kg/m2): OR = 0.60 (CI 0.56-0.65)], morbid obesity (BMI 40-49.9 kg/m2): OR = 0.29 (CI 0.23-0.37). The OR for diaphragmatic hernia peaks with obesity in women and overweight status in men but was found to decrease with morbid obesity [OR = 1.18 (CI 1.07-1.30)]. There was no significant difference between men and women in the prevalence of femoral hernia (men: 0.7/per10,000, women: 0.9/per10,000, p = 0.19). CONCLUSIONS The relationship between hernia and obesity is complex with some hernias decreasing in prevalence as obesity increases. Further research is needed to better understand this paradoxical relationship.
Collapse
Affiliation(s)
- H Zelicha
- Department of Surgery, Faculty of Health Sciences, UCLA School of Medicine, CHS 74-121, Los Angeles, CA, 90095, USA
- Department of Medicine, Division of General Internal Medicine, UCLA, Los Angeles, CA, USA
| | - D S Bell
- Department of Medicine, Division of General Internal Medicine, UCLA, Los Angeles, CA, USA
- Informatics Program of the UCLA Clinical and Translational Science Institute (CTSI), UCLA, Los Angeles, CA, USA
| | - D Chen
- Department of Surgery, Faculty of Health Sciences, UCLA School of Medicine, CHS 74-121, Los Angeles, CA, 90095, USA
| | - Y Chen
- Department of Surgery, Faculty of Health Sciences, UCLA School of Medicine, CHS 74-121, Los Angeles, CA, 90095, USA
| | - E H Livingston
- Department of Surgery, Faculty of Health Sciences, UCLA School of Medicine, CHS 74-121, Los Angeles, CA, 90095, USA.
| |
Collapse
|
23
|
DeBari MK, Johnston EK, Scott JV, Ilzuka E, Sun W, Webster-Wood VA, Abbott RD. A Preliminary Study on Factors That Drive Patient Variability in Human Subcutaneous Adipose Tissues. Cells 2024; 13:1240. [PMID: 39120271 PMCID: PMC11311805 DOI: 10.3390/cells13151240] [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: 06/10/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Adipose tissue is a dynamic regulatory organ that has profound effects on the overall health of patients. Unfortunately, inconsistencies in human adipose tissues are extensive and multifactorial, including large variability in cellular sizes, lipid content, inflammation, extracellular matrix components, mechanics, and cytokines secreted. Given the high human variability, and since much of what is known about adipose tissue is from animal models, we sought to establish correlations and patterns between biological, mechanical, and epidemiological properties of human adipose tissues. To do this, twenty-six independent variables were cataloged for twenty patients, which included patient demographics and factors that drive health, obesity, and fibrosis. A factorial analysis for mixed data (FAMD) was used to analyze patterns in the dataset (with BMI > 25), and a correlation matrix was used to identify interactions between quantitative variables. Vascular endothelial growth factor A (VEGFA) and actin alpha 2, smooth muscle (ACTA2) gene expression were the highest loadings in the first two dimensions of the FAMD. The number of adipocytes was also a key driver of patient-related differences, where a decrease in the density of adipocytes was associated with aging. Aging was also correlated with a decrease in overall lipid percentage of subcutaneous tissue, with lipid deposition being favored extracellularly, an increase in transforming growth factor-β1 (TGFβ1), and an increase in M1 macrophage polarization. An important finding was that self-identified race contributed to variance between patients in this study, where Black patients had significantly lower gene expression levels of TGFβ1 and ACTA2. This finding supports the urgent need to account for patient ancestry in biomedical research to develop better therapeutic strategies for all patients. Another important finding was that TGFβ induced factor homeobox 1 (TGIF1), an understudied signaling molecule, which is highly correlated with leptin signaling, was correlated with metabolic inflammation. Furthermore, this study draws attention to what we define as "extracellular lipid droplets", which were consistently found in collagen-rich regions of the obese adipose tissues evaluated here. Reduced levels of TGIF1 were correlated with higher numbers of extracellular lipid droplets and an inability to suppress fibrotic changes in adipose tissue. Finally, this study indicated that M1 and M2 macrophage markers were correlated with each other and leptin in patients with a BMI > 25. This finding supports growing evidence that macrophage polarization in obesity involves a complex, interconnecting network system rather than a full switch in activation patterns from M2 to M1 with increasing body mass. Overall, this study reinforces key findings in animal studies and identifies important areas for future research, where human and animal studies are divergent. Understanding key drivers of human patient variability is required to unravel the complex metabolic health of unique patients.
Collapse
Affiliation(s)
- Megan K. DeBari
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Elizabeth K. Johnston
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Jacqueline V. Scott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Erica Ilzuka
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Wenhuan Sun
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Victoria A. Webster-Wood
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Rosalyn D. Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| |
Collapse
|
24
|
Wareing N, Mills TW, Collum S, Wu M, Revercomb L, Girard R, Lyons M, Skaug B, Bi W, Ali MA, Koochak H, Flores AR, Yang Y, Zheng WJ, Swindell WR, Assassi S, Karmouty-Quintana H. Deletion of adipocyte Sine Oculis Homeobox Homolog 1 prevents lipolysis and attenuates skin fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595271. [PMID: 38826482 PMCID: PMC11142148 DOI: 10.1101/2024.05.22.595271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Dermal fibrosis is a cardinal feature of systemic sclerosis (SSc) for which there are limited treatment strategies. This is in part due to our fragmented understanding of how dermal white adipose tissue (DWAT) contributes to skin fibrosis. We identified elevated sine oculis homeobox homolog 1 (SIX1) expression in SSc skin samples from the GENISOS and PRESS cohorts, the expression of which correlated with adipose-associated genes and molecular pathways. SIX1 localization studies identified increased signals in the DWAT area in SSc and in experimental models of skin fibrosis. Global and adipocyte specific Six1 deletion abrogated end-stage fibrotic gene expression and dermal adipocyte shrinkage induced by SQ bleomycin treatment. Further studies revealed a link between elevated SIX1 and increased expression of SERPINE1 and its protein PAI-1 which are known pro-fibrotic mediators. However, SIX1 deletion did not appear to affect cellular trans differentiation. Taken together these results point at SIX1 as a potential target for dermal fibrosis in SSc.
Collapse
Affiliation(s)
- Nancy Wareing
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Tingting W Mills
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Scott Collum
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Minghua Wu
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | | | - Rene Girard
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Marka Lyons
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Brian Skaug
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Weizhen Bi
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Meer A. Ali
- D Bradley McWilliams School of Biomedical Informatics, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Haniyeh Koochak
- Department of Pediatrics, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Anthony R Flores
- Department of Pediatrics, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Yuntao Yang
- D Bradley McWilliams School of Biomedical Informatics, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - W Jim Zheng
- D Bradley McWilliams School of Biomedical Informatics, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - William R Swindell
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Shervin Assassi
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
- Divisions of Critical Care, Pulmonary and Sleep Medicine, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| |
Collapse
|
25
|
Savulescu-Fiedler I, Mihalcea R, Dragosloveanu S, Scheau C, Baz RO, Caruntu A, Scheau AE, Caruntu C, Benea SN. The Interplay between Obesity and Inflammation. Life (Basel) 2024; 14:856. [PMID: 39063610 PMCID: PMC11277997 DOI: 10.3390/life14070856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is an important condition affecting the quality of life of numerous patients and increasing their associated risk for multiple diseases, including tumors and immune-mediated disorders. Inflammation appears to play a major role in the development of obesity and represents a central point for the activity of cellular and humoral components in the adipose tissue. Macrophages play a key role as the main cellular component of the adipose tissue regulating the chronic inflammation and modulating the secretion and differentiation of various pro- and anti-inflammatory cytokines. Inflammation also involves a series of signaling pathways that might represent the focus for new therapies and interventions. Weight loss is essential in decreasing cardiometabolic risks and the degree of associated inflammation; however, the latter can persist for long after the excess weight is lost, and can involve changes in macrophage phenotypes that can ensure the metabolic adjustment. A clear understanding of the pathophysiological processes in the adipose tissue and the interplay between obesity and chronic inflammation can lead to a better understanding of the development of comorbidities and may ensure future targets for the treatment of obesity.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Razvan Mihalcea
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Radu Octavian Baz
- Clinical Laboratory of Radiology and Medical Imaging, “Sf. Apostol Andrei” County Emergency Hospital, 900591 Constanta, Romania
- Department of Radiology and Medical Imaging, Faculty of Medicine, “Ovidius” University, 900527 Constanta, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- “Prof. Dr. Matei Balș” National Institute for Infectious Diseases, 021105 Bucharest, Romania
| |
Collapse
|
26
|
Hu J, Zheng S, Hua M, Ding M, Hu Z, Jiang H. Deletion of Impdh2 in adipocyte precursors limits the expansion of white adipose tissue and enhances metabolic health with overnutrition. Biochem Biophys Res Commun 2024; 716:149998. [PMID: 38692012 DOI: 10.1016/j.bbrc.2024.149998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
The equilibrium between the hypertrophic growth of existing adipocytes and adipogenesis is vital in managing metabolic stability in white adipocytes when faced with overnutrition. Adipogenesis has been established as a key player in combating metabolic irregularities caused by various factors. However, the benefits of increasing adipogenesis-mediated white adipose tissue (WAT) expansion for metabolic health regulation remain uncertain. Our findings reveal an increase in Impdh2 expression during the adipogenesis phase, both in vivo and in vitro. Xmp enhances adipogenic potential by fostering mitotic clonal expansion (MCE). The conditional knockout of Impdh2 in adipocyte progenitor cells(APCs) in adult and aged mice effectively curbs white adipose tissue expansion, ameliorates glucose tolerance, and augments energy expenditure under high-fat diet (HFD). However, no significant difference is observed under normal chow diet (NCD). Concurrently, the knockout of Impdh2 in APCs significantly reduces the count of new adipocytes induced by HFD, without affecting adipocyte size. Mechanistically, Impdh2 regulates the proliferation of APCs during the MCE phase via Xmp. Exogenous Xmp can significantly offset the reduction in adipogenic abilities of APCs due to Impdh2 deficiency. In summary, we discovered that adipogenesis-mediated WAT expansion, induced by overnutrition, also contributes to metabolic abnormalities. Moreover, the pivotal role of Impdh2 in regulating adipogenesis in APCs offers a novel therapeutic approach to combat obesity.
Collapse
Affiliation(s)
- Jiaqi Hu
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Shiqiong Zheng
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Mengting Hua
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Mei Ding
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Zhangfeng Hu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Bio-medical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China; Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China.
| | - Haochen Jiang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Bio-medical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China.
| |
Collapse
|
27
|
Dahdah N, Tercero-Alcázar C, Malagón MM, Garcia-Roves PM, Guzmán-Ruiz R. Interrelation of adipose tissue macrophages and fibrosis in obesity. Biochem Pharmacol 2024; 225:116324. [PMID: 38815633 DOI: 10.1016/j.bcp.2024.116324] [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: 01/09/2024] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Obesity is characterized by adipose tissue expansion, extracellular matrix remodelling and unresolved inflammation that contribute to insulin resistance and fibrosis. Adipose tissue macrophages represent the most abundant class of immune cells in adipose tissue inflammation and could be key mediators of adipocyte dysfunction and fibrosis in obesity. Although macrophage activation states are classically defined by the M1/M2 polarization nomenclature, novel studies have revealed a more complex range of macrophage phenotypes in response to external condition or the surrounding microenvironment. Here, we discuss the plasticity of adipose tissue macrophages (ATMs) in response to their microenvironment in obesity, with special focus on macrophage infiltration and polarization, and their contribution to adipose tissue fibrosis. A better understanding of the role of ATMs as regulators of adipose tissue remodelling may provide novel therapeutic strategies against obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Norma Dahdah
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Carmen Tercero-Alcázar
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María M Malagón
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Cell Biology, Physiology and Immunology, IMIBIC, Reina Sofía University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Pablo Miguel Garcia-Roves
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Rocío Guzmán-Ruiz
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Cell Biology, Physiology and Immunology, IMIBIC, Reina Sofía University Hospital, University of Córdoba, 14004 Córdoba, Spain.
| |
Collapse
|
28
|
Reese-Petersen AL, Holm Nielsen S, Bülow Sand JM, Schattenberg JM, Bugianesi E, Karsdal MA. The sclerotic component of metabolic syndrome: Fibroblast activities may be the central common denominator driving organ function loss and death. Diabetes Obes Metab 2024; 26:2554-2566. [PMID: 38699780 DOI: 10.1111/dom.15615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024]
Abstract
Fibrosis is a common feature of more than 50 different diseases and the cause of more than 35% of deaths worldwide, of which liver, kidney, skin, heart and, recently, lungs are receiving the most attention. Tissue changes, resulting in loss of organ function, are both a cause and consequence of disease and outcome. Fibrosis is caused by an excess deposition of extracellular matrix proteins, which over time results in impaired organ function and organ failure, and the pathways leading to increased fibroblast activation are many. This narrative review investigated the common denominator of fibrosis, fibroblasts, and the activation of fibroblasts, in response to excess energy consumption in liver, kidney, heart, skin and lung fibrosis. Fibroblasts are the main drivers of organ function loss in lung, liver, skin, heart and kidney disease. Fibroblast activation in response to excess energy consumption results in the overproduction of a range of collagens, of which types I, III and VI seem to be the essential drivers of disease progression. Fibroblast activation may be quantified in serum, enabling profiling and selection of patients. Activation of fibroblasts results in the overproduction of collagens, which deteriorates organ function. Patient profiling of fibroblast activities in serum, quantified as collagen production, may identify an organ death trajectory, better enabling identification of the right treatment for use in different metabolic interventions. As metabolically activated patients have highly elevated risk of kidney, liver and heart failure, it is essential to identify which organ to treat first and monitor organ status to correct treatment regimes. In direct alignment with this, it is essential to identify the right patients with the right organ deterioration trajectory for enrolment in clinical studies.
Collapse
Affiliation(s)
| | | | | | - Jörn M Schattenberg
- Saarland University Medical Center, Homburg, Germany
- University of the Saarland, Saarbrücken, Germany
| | | | | |
Collapse
|
29
|
Blade SP, Falkowski DJ, Bachand SN, Pagano SJ, Chin L. Mechanobiology of Adipocytes. BIOLOGY 2024; 13:434. [PMID: 38927314 PMCID: PMC11200640 DOI: 10.3390/biology13060434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024]
Abstract
The growing obesity epidemic necessitates increased research on adipocyte and adipose tissue function and disease mechanisms that progress obesity. Historically, adipocytes were viewed simply as storage for excess energy. However, recent studies have demonstrated that adipocytes play a critical role in whole-body homeostasis, are involved in cell communication, experience forces in vivo, and respond to mechanical stimuli. Changes to the adipocyte mechanical microenvironment can affect function and, in some cases, contribute to disease. The aim of this review is to summarize the current literature on the mechanobiology of adipocytes. We reviewed over 100 papers on how mechanical stress is sensed by the adipocyte, the effects on cell behavior, and the use of cell culture scaffolds, particularly those with tunable stiffness, to study adipocyte behavior, adipose cell and tissue mechanical properties, and computational models. From our review, we conclude that adipocytes are responsive to mechanical stimuli, cell function and adipogenesis can be dictated by the mechanical environment, the measurement of mechanical properties is highly dependent on testing methods, and current modeling practices use many different approaches to recapitulate the complex behavior of adipocytes and adipose tissue. This review is intended to aid future studies by summarizing the current literature on adipocyte mechanobiology.
Collapse
Affiliation(s)
- Sean P. Blade
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Dylan J. Falkowski
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Sarah N. Bachand
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Steven J. Pagano
- Department of Mechanical Engineering, Widener University, Chester, PA 19013, USA;
| | - LiKang Chin
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| |
Collapse
|
30
|
Henriksen K, Genovese F, Reese-Petersen A, Audoly LP, Sun K, Karsdal MA, Scherer PE. Endotrophin, a Key Marker and Driver for Fibroinflammatory Disease. Endocr Rev 2024; 45:361-378. [PMID: 38091968 PMCID: PMC11492497 DOI: 10.1210/endrev/bnad036] [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: 08/08/2023] [Revised: 11/02/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024]
Abstract
Our overview covers several key areas related to recent results obtained for collagen type VI and endotrophin (ETP). (1) An introduction to the history of ETP, including how it was identified, how it is released, and its function and potential receptors. (2) An introduction to the collagen family, with a focus on what differentiates collagen type VI from an evolutionary standpoint. (3) An overview of collagen type VI, the 6 individual chains (COL6A1, A2, A3, A4, A5, and A6), their differences and similarities, as well as their expression profiles and function. (4) A detailed analysis of COL6A3, including the cleaved product endotrophin, and what separates it from the other 5 collagen 6 molecules, including its suggested function based on insights gained from knockout and gain of function mouse models. (5) The pathology of ETP. What leads to its presence and release and what are the consequences thereof? (6) Functional implications of circulating ETP. Here we review the data with the functional roles of ETP in mind. (7) We propose that ETP is a mediator for fibrotic (or fibroinflammatory) disorders. Based on what we know about ETP, we have to consider it as a target for the treatment of fibrotic (or fibroinflammatory) disorders. What segment(s) of the patient population would most dramatically respond to an ETP-targeted intervention? How can we find the population that would profit most from an intervention? We aim to present a broad overview over the ETP field at large, providing an assessment of where the future research efforts need to be placed to tap into the vast potential of ETP, both as a marker and as a target in different diseases.
Collapse
Affiliation(s)
- Kim Henriksen
- Department of Cardiovascular Disease, Nordic Bioscience A/S, DK-2730 Herlev, Denmark
| | - Federica Genovese
- Department of Cardiovascular Disease, Nordic Bioscience A/S, DK-2730 Herlev, Denmark
| | | | | | - Kai Sun
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Morten A Karsdal
- Department of Cardiovascular Disease, Nordic Bioscience A/S, DK-2730 Herlev, Denmark
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
31
|
Drygalski K, Higos R, Merabtene F, Mojsak P, Grubczak K, Ciborowski M, Razak H, Clément K, Dugail I. Extracellular matrix hyaluronan modulates fat cell differentiation and primary cilia dynamics. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159470. [PMID: 38423452 DOI: 10.1016/j.bbalip.2024.159470] [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: 12/04/2023] [Revised: 02/02/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
Hyaluronan is an important extracellular matrix component, with poorly documented physiological role in the context of lipid-rich adipose tissue. We have investigated the global impact of hyaluronan removal from adipose tissue environment by in vitro exposure to exogenous hyaluronidase (or heat inactivated enzyme). Gene set expression analysis from RNA sequencing revealed downregulated adipogenesis as a main response to hyaluronan removal from human adipose tissue samples, which was confirmed by hyaluronidase-mediated inhibition of adipocyte differentiation in the 3T3L1 adipose cell line. Hyaluronidase exposure starting from the time of induction with the differentiation cocktail reduced lipid accumulation in mature adipocytes, limited the expression of terminal differentiation marker genes, and impaired the early induction of co-regulated Cebpa and Pparg mRNA. Reduction of Cebpa and Pparg expression by exogenous hyaluronidase was also observed in cultured primary preadipocytes from subcutaneous, visceral or brown adipose tissue of mice. Mechanistically, inhibition of adipogenesis by hyaluronan removal was not caused by changes in osmotic pressure or cell inflammatory status, could not be mimicked by exposure to threose, a metabolite generated by hyaluronan degradation, and was not linked to alteration in endogenous Wnt ligands expression. Rather, we observed that hyaluronan removal associated with disrupted primary cilia dynamics, with elongated cilium and higher proportions of preadipocytes that remained ciliated in hyaluronidase-treated conditions. Thus, our study points to a new link between ciliogenesis and hyaluronan impacting adipose tissue development.
Collapse
Affiliation(s)
- Krzysztof Drygalski
- INSERM, Sorbonne Université, NutriOmics team : Nutrition/Obesities- systemic approaches, Paris 75013, France; Department of Hypertension and Diabetology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Romane Higos
- INSERM, Sorbonne Université, NutriOmics team : Nutrition/Obesities- systemic approaches, Paris 75013, France
| | - Fatiha Merabtene
- INSERM, Sorbonne Université, NutriOmics team : Nutrition/Obesities- systemic approaches, Paris 75013, France
| | - Patrycja Mojsak
- Clinical Research Centre, Medical University of Bialystok, 15-276 Białystok, Poland
| | - Kamil Grubczak
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Michal Ciborowski
- Clinical Research Centre, Medical University of Bialystok, 15-276 Białystok, Poland
| | - Hady Razak
- Department of General and Endocrine Surgery, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Karine Clément
- INSERM, Sorbonne Université, NutriOmics team : Nutrition/Obesities- systemic approaches, Paris 75013, France; Assistance Publique-Hopitaux de Paris, Nutrition department, Pitié-Salpetrière Hospital, 75013 Paris, France
| | - Isabelle Dugail
- INSERM, Sorbonne Université, NutriOmics team : Nutrition/Obesities- systemic approaches, Paris 75013, France.
| |
Collapse
|
32
|
Zhu X, Zeng C, Yu B. White adipose tissue in metabolic associated fatty liver disease. Clin Res Hepatol Gastroenterol 2024; 48:102336. [PMID: 38604293 DOI: 10.1016/j.clinre.2024.102336] [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: 12/05/2023] [Revised: 03/28/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Metabolic associated fatty liver disease (MAFLD) is a prevalent chronic liver condition globally, currently lacking universally recognized therapeutic drugs, thereby increasing the risk of cirrhosis and hepatocellular carcinoma. Research has reported an association between white adipose tissue and MAFLD. SCOPE OF REVIEW White adipose tissue (WAT) is involved in lipid metabolism and can contribute to the progression of MAFLD by mediating insulin resistance, inflammation, exosomes, autophagy, and other processes. This review aims to elucidate the mechanisms through which WAT plays a role in the development of MAFLD. MAJOR CONCLUSIONS WAT participates in the occurrence and progression of MAFLD by mediating insulin resistance, inflammation, autophagy, and exosome secretion. Fibrosis and restricted expansion of adipose tissue can lead to the release of more free fatty acids (FFA), exacerbating the progression of MAFLD. WAT-secreted TNF-α and IL-1β, through the promotion of JNK/JKK/p38MAPK expression, interfere with insulin receptor serine and tyrosine phosphorylation, worsening insulin resistance. Adiponectin, by inhibiting the TLR-4-NF-κB pathway and suppressing M2 to M1 transformation, further inhibits the secretion of IL-6, IL-1β, and TNF-α, improving insulin resistance in MAFLD patients. Various gene expressions within WAT, such as MBPAT7, Nrf2, and Ube4A, can ameliorate insulin resistance in MAFLD patients. Autophagy-related gene Atg7 promotes the expression of fibrosis-related genes, worsening MAFLD. Non-pharmacological treatments, including diabetes-related medications and exercise, can improve MAFLD.
Collapse
Affiliation(s)
- Xiaoqin Zhu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, Hubei, 430000, PR China
| | - Chuanfei Zeng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, Hubei, 430000, PR China
| | - Baoping Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, Hubei, 430000, PR China.
| |
Collapse
|
33
|
Hateley C, Olona A, Halliday L, Edin ML, Ko JH, Forlano R, Terra X, Lih FB, Beltrán-Debón R, Manousou P, Purkayastha S, Moorthy K, Thursz MR, Zhang G, Goldin RD, Zeldin DC, Petretto E, Behmoaras J. Multi-tissue profiling of oxylipins reveal a conserved up-regulation of epoxide:diol ratio that associates with white adipose tissue inflammation and liver steatosis in obesity. EBioMedicine 2024; 103:105127. [PMID: 38677183 PMCID: PMC11061246 DOI: 10.1016/j.ebiom.2024.105127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Obesity drives maladaptive changes in the white adipose tissue (WAT) which can progressively cause insulin resistance, type 2 diabetes mellitus (T2DM) and metabolic dysfunction-associated liver disease (MASLD). Obesity-mediated loss of WAT homeostasis can trigger liver steatosis through dysregulated lipid pathways such as those related to polyunsaturated fatty acid (PUFA)-derived oxylipins. However, the exact relationship between oxylipins and metabolic syndrome remains elusive and cross-tissue dynamics of oxylipins are ill-defined. METHODS We quantified PUFA-related oxylipin species in the omental WAT, liver biopsies and plasma of 88 patients undergoing bariatric surgery (female N = 79) and 9 patients (female N = 4) undergoing upper gastrointestinal surgery, using UPLC-MS/MS. We integrated oxylipin abundance with WAT phenotypes (adipogenesis, adipocyte hypertrophy, macrophage infiltration, type I and VI collagen remodelling) and the severity of MASLD (steatosis, inflammation, fibrosis) quantified in each biopsy. The integrative analysis was subjected to (i) adjustment for known risk factors and, (ii) control for potential drug-effects through UPLC-MS/MS analysis of metformin-treated fat explants ex vivo. FINDINGS We reveal a generalized down-regulation of cytochrome P450 (CYP)-derived diols during obesity conserved between the WAT and plasma. Notably, epoxide:diol ratio, indicative of soluble epoxide hydrolyse (sEH) activity, increases with WAT inflammation/fibrosis, hepatic steatosis and T2DM. Increased 12,13-EpOME:DiHOME in WAT and liver is a marker of worsening metabolic syndrome in patients with obesity. INTERPRETATION These findings suggest a dampened sEH activity and a possible role of fatty acid diols during metabolic syndrome in major metabolic organs such as WAT and liver. They also have implications in view of the clinical trials based on sEH inhibition for metabolic syndrome. FUNDING Wellcome Trust (PS3431_WMIH); Duke-NUS (Intramural Goh Cardiovascular Research Award (Duke-NUS-GCR/2022/0020); National Medical Research Council (OFLCG22may-0011); National Institute of Environmental Health Sciences (Z01 ES025034); NIHR Imperial Biomedical Research Centre.
Collapse
Affiliation(s)
- Charlotte Hateley
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Antoni Olona
- Centre for Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Laura Halliday
- Department of Surgery and Cancer, Imperial College London, UK
| | - Matthew L Edin
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Jeong-Hun Ko
- Division of Brain Sciences, Imperial College Faculty of Medicine, London, UK
| | - Roberta Forlano
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Ximena Terra
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, MoBioFood Research Group, Tarragona, Spain
| | - Fred B Lih
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Raúl Beltrán-Debón
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, MoBioFood Research Group, Tarragona, Spain
| | - Penelopi Manousou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Sanjay Purkayastha
- Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK; University of Brunel, Kingston Lane, Uxbridge, London, UB8 3PH, UK
| | - Krishna Moorthy
- Department of Surgery and Cancer, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Mark R Thursz
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Guodong Zhang
- Department of Nutrition, College of Agriculture and Environmental Sciences, 3135 Meyer Hall, One Shields Avenue, UC Davis, Davis, CA, 95616, USA
| | - Robert D Goldin
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Darryl C Zeldin
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Enrico Petretto
- Centre for Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore; Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University (CPU), Nanjing, China
| | - Jacques Behmoaras
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK; Centre for Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
34
|
Gaebler D, Hachey SJ, Hughes CCW. Microphysiological systems as models for immunologically 'cold' tumors. Front Cell Dev Biol 2024; 12:1389012. [PMID: 38711620 PMCID: PMC11070549 DOI: 10.3389/fcell.2024.1389012] [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: 02/20/2024] [Accepted: 03/25/2024] [Indexed: 05/08/2024] Open
Abstract
The tumor microenvironment (TME) is a diverse milieu of cells including cancerous and non-cancerous cells such as fibroblasts, pericytes, endothelial cells and immune cells. The intricate cellular interactions within the TME hold a central role in shaping the dynamics of cancer progression, influencing pivotal aspects such as tumor initiation, growth, invasion, response to therapeutic interventions, and the emergence of drug resistance. In immunologically 'cold' tumors, the TME is marked by a scarcity of infiltrating immune cells, limited antigen presentation in the absence of potent immune-stimulating signals, and an abundance of immunosuppressive factors. While strategies targeting the TME as a therapeutic avenue in 'cold' tumors have emerged, there is a pressing need for novel approaches that faithfully replicate the complex cellular and non-cellular interactions in order to develop targeted therapies that can effectively stimulate immune responses and improve therapeutic outcomes in patients. Microfluidic devices offer distinct advantages over traditional in vitro 3D co-culture models and in vivo animal models, as they better recapitulate key characteristics of the TME and allow for precise, controlled insights into the dynamic interplay between various immune, stromal and cancerous cell types at any timepoint. This review aims to underscore the pivotal role of microfluidic systems in advancing our understanding of the TME and presents current microfluidic model systems that aim to dissect tumor-stromal, tumor-immune and immune-stromal cellular interactions in various 'cold' tumors. Understanding the intricacies of the TME in 'cold' tumors is crucial for devising effective targeted therapies to reinvigorate immune responses and overcome the challenges of current immunotherapy approaches.
Collapse
Affiliation(s)
- Daniela Gaebler
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Stephanie J. Hachey
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Christopher C. W. Hughes
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
- Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
35
|
Park S, Shimokawa I. Influence of Adipokines on Metabolic Dysfunction and Aging. Biomedicines 2024; 12:873. [PMID: 38672227 PMCID: PMC11048512 DOI: 10.3390/biomedicines12040873] [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: 03/19/2024] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Currently, 30% of the global population is overweight or obese, with projections from the World Obesity Federation suggesting that this figure will surpass 50% by 2035. Adipose tissue dysfunction, a primary characteristic of obesity, is closely associated with an increased risk of metabolic abnormalities, such as hypertension, hyperglycemia, and dyslipidemia, collectively termed metabolic syndrome. In particular, visceral fat accretion is considered as a hallmark of aging and is strongly linked to higher mortality rates in humans. Adipokines, bioactive peptides secreted by adipose tissue, play crucial roles in regulating appetite, satiety, adiposity, and metabolic balance, thereby rendering them key players in alleviating metabolic diseases and potentially extending health span. In this review, we elucidated the role of adipokines in the development of obesity and related metabolic disorders while also exploring the potential of certain adipokines as candidates for longevity interventions.
Collapse
Affiliation(s)
- Seongjoon Park
- Department of Pathology, Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan;
| | - Isao Shimokawa
- Department of Pathology, Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan;
- SAGL, Limited Liability Company, 1-4-34, Kusagae, Chuo-ku, Fukuoka 810-0045, Japan
| |
Collapse
|
36
|
Caballero-Sánchez N, Alonso-Alonso S, Nagy L. Regenerative inflammation: When immune cells help to re-build tissues. FEBS J 2024; 291:1597-1614. [PMID: 36440547 PMCID: PMC10225019 DOI: 10.1111/febs.16693] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/29/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022]
Abstract
Inflammation is an essential immune response critical for responding to infection, injury and maintenance of tissue homeostasis. Upon injury, regenerative inflammation promotes tissue repair by a timed and coordinated infiltration of diverse cell types and the secretion of growth factors, cytokines and lipids mediators. Remarkably, throughout evolution as well as mammalian development, this type of physiological inflammation is highly associated with immunosuppression. For instance, regenerative inflammation is the consequence of an in situ macrophage polarization resulting in a transition from pro-inflammatory to anti-inflammatory/pro-regenerative response. Immune cells are the first responders upon injury, infiltrating the damaged tissue and initiating a pro-inflammatory response depleting cell debris and necrotic cells. After phagocytosis, macrophages undergo multiple coordinated metabolic and transcriptional changes allowing the transition and dictating the initiation of the regenerative phase. Differences between a highly efficient, complete ad integrum tissue repair, such as, acute skeletal muscle injury, and insufficient regenerative inflammation, as the one developing in Duchenne Muscular Dystrophy (DMD), highlight the importance of a coordinated response orchestrated by immune cells. During regenerative inflammation, these cells interact with others and alter the niche, affecting the character of inflammation itself and, therefore, the progression of tissue repair. Comparing acute muscle injury and chronic inflammation in DMD, we review how the same cells and molecules in different numbers, concentration and timing contribute to very different outcomes. Thus, it is important to understand and identify the distinct functions and secreted molecules of macrophages, and potentially other immune cells, during tissue repair, and the contributors to the macrophage switch leveraging this knowledge in treating diseases.
Collapse
Affiliation(s)
- Noemí Caballero-Sánchez
- Doctoral School of Molecular Cell and Immunobiology, Faculty of Medicine, University of Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
| | - Sergio Alonso-Alonso
- Instituto Oftalmológico Fernández-Vega, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Laszlo Nagy
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
- Departments Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| |
Collapse
|
37
|
Hagberg CE, Spalding KL. White adipocyte dysfunction and obesity-associated pathologies in humans. Nat Rev Mol Cell Biol 2024; 25:270-289. [PMID: 38086922 DOI: 10.1038/s41580-023-00680-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 02/10/2024]
Abstract
The prevalence of obesity and associated chronic diseases continues to increase worldwide, negatively impacting on societies and economies. Whereas the association between excess body weight and increased risk for developing a multitude of diseases is well established, the initiating mechanisms by which weight gain impairs our metabolic health remain surprisingly contested. In order to better address the myriad of disease states associated with obesity, it is essential to understand adipose tissue dysfunction and develop strategies for reinforcing adipocyte health. In this Review we outline the diverse physiological functions and pathological roles of human white adipocytes, examining our current knowledge of why white adipocytes are vital for systemic metabolic control, yet poorly adapted to our current obesogenic environment.
Collapse
Affiliation(s)
- Carolina E Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kirsty L Spalding
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
38
|
Otani T, Mizokami A, Takeuchi H, Inai T, Hirata M. The role of adhesion molecules in osteocalcin-induced effects on glucose and lipid metabolism in adipocytes. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119701. [PMID: 38417588 DOI: 10.1016/j.bbamcr.2024.119701] [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: 10/09/2023] [Revised: 02/08/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
Recent findings suggest that uncarboxylated osteocalcin (GluOC) promotes glucose and lipid metabolism via its putative receptor GPRC6A; however, its direct effect on adipocytes remains elusive. In this study, we elucidated the effects of GluOC on adipocytes, with an emphasis on the role of cell adhesion molecules. We determined that GluOC promoted the expression of adipocyte adhesion molecule (ACAM) and its transcription factor Krüppel-like factor 4 and enhanced the cortical actin filament assembly, which ameliorated lipid droplet hypertrophy. Additionally, GluOC upregulated the expression of integrin αVβ3 and activation of focal adhesion kinase (FAK) and prevented insulin receptor substrate 1 (IRS1) degradation by inhibiting the ubiquitin-proteasome system via the FAK-PLC-PKC axis, which activated IRS1-Akt-mediated glucose transporter 4 (GLUT4) transport. Furthermore, we showed that GluOC elevated the expression of the insulin-independent glucose transporters GLUT1 and GLUT8, which facilitated insulin stimulation-independent glucose transport. The GluOC-induced activation of integrin αVβ3 signaling promoted microtubule assembly, which improved glucose and lipid metabolism via its involvement in intracellular vesicular transport. GluOC treatment also suppressed collagen type 1 formation, which might prevent adipose tissue fibrosis in obese individuals. Overall, our results imply that GluOC promotes glucose and lipid metabolism via ACAM, integrin αVβ3, and GLUT1 and 8 expression, directly affecting adipocytes.
Collapse
Affiliation(s)
- Takahito Otani
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka 814-0193, Japan.
| | - Akiko Mizokami
- Oral Health/Brain Health/Total Health Research Center, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Hiroshi Takeuchi
- Division of Applied Pharmacology, Kyushu Dental University, Kitakyushu 803-8580, Japan
| | - Tetsuichiro Inai
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka 814-0193, Japan
| | - Masato Hirata
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka 814-0193, Japan.
| |
Collapse
|
39
|
Zhou XT, Zhu AQ, Li XM, Sun LY, Yan JG, Luo N, Chen SS, Huang Z, Mao XL, Li KP. Mulberry and Hippophae-based solid beverage promotes weight loss in rats by antagonizing white adipose tissue PPARγ and FGFR1 signaling. Front Endocrinol (Lausanne) 2024; 15:1344262. [PMID: 38559696 PMCID: PMC10978776 DOI: 10.3389/fendo.2024.1344262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Obesity, a multifactorial disease with many complications, has become a global epidemic. Weight management, including dietary supplementation, has been confirmed to provide relevant health benefits. However, experimental evidence and mechanistic elucidation of dietary supplements in this regard are limited. Here, the weight loss efficacy of MHP, a commercial solid beverage consisting of mulberry leaf aqueous extract and Hippophae protein peptides, was evaluated in a high-fat high-fructose (HFF) diet-induced rat model of obesity. Body component analysis and histopathologic examination confirmed that MHP was effective to facilitate weight loss and adiposity decrease. Pathway enrichment analysis with differential metabolites generated by serum metabolomic profiling suggests that PPAR signal pathway was significantly altered when the rats were challenged by HFF diet but it was rectified after MHP intervention. RNA-Seq based transcriptome data also indicates that MHP intervention rectified the alterations of white adipose tissue mRNA expressions in HFF-induced obese rats. Integrated omics reveals that the efficacy of MHP against obesogenic adipogenesis was potentially associated with its regulation of PPARγ and FGFR1 signaling pathway. Collectively, our findings suggest that MHP could improve obesity, providing an insight into the use of MHP in body weight management.
Collapse
Affiliation(s)
- Xiao-Ting Zhou
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - An-Qi Zhu
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiao-Min Li
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Ling-Yue Sun
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jian-Gang Yan
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Nin Luo
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shi-Sheng Chen
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Zebo Huang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Xin-Liang Mao
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Kun-Ping Li
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
40
|
Sawamoto A, Okada M, Matsuoka N, Okuyama S, Nakajima M. Tipepidine activates AMPK and improves adipose tissue fibrosis and glucose intolerance in high-fat diet-induced obese mice. FASEB J 2024; 38:e23542. [PMID: 38466234 DOI: 10.1096/fj.202301861rr] [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: 09/12/2023] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/12/2024]
Abstract
Tipepidine (3-[di-2-thienylmethylene]-1-methylpiperidine) (TP) is a non-narcotic antitussive used in Japan. Recently, the potential application of TP in the treatment of neuropsychiatric disorders, such as depression and attention deficit hyperactivity disorder, has been suggested; however, its functions in energy metabolism are unknown. Here, we demonstrate that TP exhibits a metabolism-improving action. The administration of TP reduced high-fat diet-induced body weight gain in mice and lipid accumulation in the liver and increased the weight of epididymal white adipose tissue (eWAT) in diet-induced obese (DIO) mice. Furthermore, TP inhibited obesity-induced fibrosis in the eWAT. We also found that TP induced AMP-activated protein kinase (AMPK) activation in the eWAT of DIO mice and 3T3-L1 cells. TP-induced AMPK activation was abrogated by the transfection of liver kinase B1 siRNA in 3T3-L1 cells. The metabolic effects of TP were almost equivalent to those of metformin, an AMPK activator that is used as a first-line antidiabetic drug. In summary, TP is a potent AMPK activator, suggesting its novel role as an antidiabetic drug owing to its antifibrotic effect on adipose tissues.
Collapse
Affiliation(s)
- Atsushi Sawamoto
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime, Japan
| | - Madoka Okada
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime, Japan
| | - Nanako Matsuoka
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime, Japan
| | - Satoshi Okuyama
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime, Japan
| | - Mitsunari Nakajima
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime, Japan
| |
Collapse
|
41
|
Conte C, Cipponeri E, Roden M. Diabetes Mellitus, Energy Metabolism, and COVID-19. Endocr Rev 2024; 45:281-308. [PMID: 37934800 PMCID: PMC10911957 DOI: 10.1210/endrev/bnad032] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/30/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023]
Abstract
Obesity, diabetes mellitus (mostly type 2), and COVID-19 show mutual interactions because they are not only risk factors for both acute and chronic COVID-19 manifestations, but also because COVID-19 alters energy metabolism. Such metabolic alterations can lead to dysglycemia and long-lasting effects. Thus, the COVID-19 pandemic has the potential for a further rise of the diabetes pandemic. This review outlines how preexisting metabolic alterations spanning from excess visceral adipose tissue to hyperglycemia and overt diabetes may exacerbate COVID-19 severity. We also summarize the different effects of SARS-CoV-2 infection on the key organs and tissues orchestrating energy metabolism, including adipose tissue, liver, skeletal muscle, and pancreas. Last, we provide an integrative view of the metabolic derangements that occur during COVID-19. Altogether, this review allows for better understanding of the metabolic derangements occurring when a fire starts from a small flame, and thereby help reducing the impact of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Caterina Conte
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome 00166, Italy
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan 20099, Italy
| | - Elisa Cipponeri
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan 20099, Italy
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Neuherberg 85764, Germany
| |
Collapse
|
42
|
Eisinger K, Girke P, Buechler C, Krautbauer S. Adipose tissue depot specific expression and regulation of fibrosis-related genes and proteins in experimental obesity. Mamm Genome 2024; 35:13-30. [PMID: 37884762 PMCID: PMC10884164 DOI: 10.1007/s00335-023-10022-3] [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: 10/14/2022] [Accepted: 10/02/2023] [Indexed: 10/28/2023]
Abstract
Transforming growth factor beta (Tgfb) is a well-studied pro-fibrotic cytokine, which upregulates cellular communication network factor 2 (Ccn2), collagen, and actin alpha 2, smooth muscle (Acta2) expression. Obesity induces adipose tissue fibrosis, which contributes to metabolic diseases. This work aimed to analyze the expression of Tgfb, Ccn2, collagen1a1 (Col1a1), Acta2 and BMP and activin membrane-bound inhibitor (Bambi), which is a negative regulator of Tgfb signaling, in different adipose tissue depots of mice fed a standard chow, mice fed a high fat diet (HFD) and ob/ob mice. Principally, these genes were low expressed in brown adipose tissues and this difference was less evident for the ob/ob mice. Ccn2 and Bambi protein as well as mRNA expression, and collagen1a1 mRNA were not induced in the adipose tissues upon HFD feeding whereas Tgfb and Acta2 mRNA increased in the white fat depots. Immunoblot analysis showed that Acta2 protein was higher in subcutaneous and perirenal fat of these mice. In the ob/ob mice, Ccn2 mRNA and Ccn2 protein were upregulated in the fat depots. Here, Tgfb, Acta2 and Col1a1 mRNA levels and serum Tgfb protein were increased. Acta2 protein was, however, not higher in subcutaneous and perirenal fat of these mice. Col6a1 mRNA was shown before to be higher in obese fat tissues. Current analysis proved the Col6a1 protein was induced in subcutaneous fat of HFD fed mice. Notably, Col6a1 was reduced in perirenal fat of ob/ob mice in comparison to the respective controls. 3T3-L1 cells express Ccn2 and Bambi protein, whose levels were not changed by fatty acids, leptin, lipopolysaccharide, tumor necrosis factor and interleukin-6. All of these factors led to higher Tgfb in 3T3-L1 adipocyte media but did not increase its mRNA levels. Free fatty acids induced necrosis whereas apoptosis did not occur in any of the in vitro incubations excluding cell death as a main reason for higher Tgfb in cell media. In summary, Tgfb mRNA is consistently induced in white fat tissues in obesity but this is not paralleled by a clear increase of its target genes. Moreover, discrepancies between mRNA and protein expression of Acta2 were observed. Adipocytes seemingly do not contribute to higher Tgfb mRNA levels in obesity. These cells release more Tgfb protein when challenged with obesity-related metabolites connecting metabolic dysfunction and fibrosis.
Collapse
Affiliation(s)
- Kristina Eisinger
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Philipp Girke
- Department of Genetics, University of Regensburg, 93040, Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany.
| | - Sabrina Krautbauer
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| |
Collapse
|
43
|
Zhang Z, Chen H, Pan C, Li R, Zhao W, Song T. Sulforaphane reduces adipose tissue fibrosis via promoting M2 macrophages polarization in HFD fed-mice. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119626. [PMID: 37977492 DOI: 10.1016/j.bbamcr.2023.119626] [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: 05/08/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Adipose tissue fibrosis has been identified as a novel contributor to the pathomechanism of obesity associated metabolic disorders. Sulforaphane (SFN) has been shown to have an anti-obesity effect. However, the impact of SFN on adipose tissue fibrosis is still not well understood. In this study, obese mice induced by high-fat diets (HFD) were used to examine the effects of SFN on adipose tissue fibrosis. According to the current findings, SFN dramatically enhanced glucose tolerance and decreased body weight in diet-induced-obesity (DIO) mice. Additionally, SFN therapy significantly reduced extracellular matrix (ECM) deposition and altered the expression of genes related to fibrosis. Furthermore, SFN also reduced inflammation and promoted macrophages polarization towards to M2 phenotype in adipose tissue, which protected adipose tissue from fibrosis. Notably, SFN-mediated nuclear factor E2-related factor 2 (Nrf2) activation was crucial in decreasing adipose tissue fibrosis. These results implied that SFN had favorable benefits in adipose tissue fibrosis, which consequently ameliorates obesity-related metabolic problems. Our research provides new treatment strategies for obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Provence, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Huali Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Cheng Pan
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Rui Li
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Wangsheng Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China.
| | - Tianzeng Song
- Institute of Animal Science, Tibet Academy of Agricultural & Animal Husbandry Science, Lhasa 850009, China.
| |
Collapse
|
44
|
Yang YY, Qi JJ, Jiang SY, Ye L. Esculin ameliorates obesity-induced insulin resistance by improving adipose tissue remodeling and activating the IRS1/PI3K/AKT/GLUT4 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117251. [PMID: 37778516 DOI: 10.1016/j.jep.2023.117251] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/13/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cortex fraxini (also known as qinpi)-the bark of Fraxinus rhynchophylla Hance (Oleaceae)-is widely used as a Chinese traditional medicinal for its anti-inflammatory and anti-hyperuricemic activities. AIM OF THE STUDY Obesity-induced insulin resistance (IR) is driving the rising incidence of type 2 diabetes mellitus and is related to pathological adipose tissue remodeling. Esculin, a major active component of Cortex fraxini, has anti-diabetic effects. However, whether esculin improves obesity-induced IR by regulating adipose tissue remodeling is unclear. The aims of the present study were to assess the effects of esculin on obesity-induced IR and to explore the underlying mechanisms. MATERIALS AND METHODS Obese IR C57BL/6J mice were treated with esculin (40 or 80 mg/kg/day) for 4 weeks. Oral glucose tolerance tests were used to assess insulin sensitivity. Histological analyses were performed to analyze the number and size distribution of adipocytes. Glucose uptake was assessed using 2-NBDG. RESULTS Esculin had no effect on body weight gain but reduced fasting blood glucose, improved oral glucose tolerance, and increased insulin sensitivity. Esculin reduced adipocyte size and the expression levels of collagen 4A1 and tumor necrosis factor α and increased the number of adipocytes and the expression of vascular endothelial growth factor A. Esculin promoted the differentiation of 3T3-L1 cells and upregulated the mRNA expression of CCAAT/enhancer-binding protein α and peroxisome proliferator-activated receptor-γ, activated the insulin receptor substrate 1 (IRS1)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, and enhanced the translocation of glucose transporter type 4 (GLUT4) and glucose uptake in adipocytes treated with palmitic acid. CONCLUSIONS These data suggest that esculin increases insulin sensitivity by improving adipose tissue remodeling and activating the IRS1/PI3K/AKT/GLUT4 pathway.
Collapse
Affiliation(s)
- Yong-Yu Yang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Hunan Provincial Engineering Research Central of Translational Medical and Innovative Drug, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Jing-Jing Qi
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Si-Yi Jiang
- Department of Pharmacy, Medical College, Yueyang Vocational Technical College, YueYang, Hunan, China.
| | - Ling Ye
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
45
|
Engin A. Adipose Tissue Hypoxia in Obesity: Clinical Reappraisal of Hypoxia Hypothesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:329-356. [PMID: 39287857 DOI: 10.1007/978-3-031-63657-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Obese subjects exhibit lower adipose tissue oxygen consumption in accordance with the lower adipose tissue blood flow. Thereby, compared to lean subjects, obese individuals have almost half lower capillary density and more than half lower vascular endothelial growth factor (VEGF). The VEGF expression together with hypoxia-inducible transcription factor-1 alpha (HIF-1α) activity also requires phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR)-mediated signaling. Especially HIF-1α is an important signaling molecule for hypoxia to induce the inflammatory responses. Hypoxia contributes to several biological functions, such as angiogenesis, cell proliferation, apoptosis, inflammation, and insulin resistance (IR). Pathogenesis of obesity-related comorbidities is attributed to intermittent hypoxia (IH), which is mostly observed in visceral obesity. Proinflammatory phenotype of the adipose tissue is a crucial link between IH and the development of IR. Inhibition of adaptive unfolded protein response (UPR) in hypoxia increases β cell death. Moreover, deletion of HIF-1α worsens β cell function. Oxidative stress, as well as the release of proinflammatory cytokines/adipokines in obesity, is proportional to the severity of IH. Reactive oxygen species (ROS) generation at mitochondria is responsible for propagation of the hypoxic signal; however, mitochondrial ROS production is required for hypoxic HIF-1α protein stabilization. Alterations in oxygen availability of adipose tissue directly affect the macrophage polarization and are responsible for the dysregulated adipocytokines production in obesity. Hypoxia both inhibits adipocyte differentiation from preadipocytes and macrophage migration from the hypoxic adipose tissue. Upon reaching a hypertrophic threshold beyond the adipocyte fat loading capacity, excess extracellular matrix (ECM) components are deposited, causing fibrosis. HIF-1α initiates the whole pathological process of fibrosis and inflammation in the obese adipose tissue. In addition to stressed adipocytes, hypoxia contributes to immune cell migration and activation which further aggravates adipose tissue fibrosis. Therefore, targeting HIF-1α might be an efficient way to suppress hypoxia-induced pathological changes in the ECM. The fibrosis score of adipose tissue correlates negatively with the body mass index and metabolic parameters. Inducers of browning/beiging adipocytes and adipokines, as well as modulations of matrix remodeling enzyme inhibitors, and associated gene regulators, are potential pharmacological targets for treating obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
46
|
Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose Tissue Dysfunction Determines Lipotoxicity and Triggers the Metabolic Syndrome: Current Challenges and Clinical Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:231-272. [PMID: 39287854 DOI: 10.1007/978-3-031-63657-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis, and it is constituted of three different types of adipocytes: white, beige and brown, which are integrated with vascular, immune, neural, and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concerted action of the three types of adipocytes/tissues ensures an optimal metabolic status. However, when one or several of these adipose depots become dysfunctional because of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations close a vicious cycle that negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and ensuring its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity are complementary strategies that counteract obesity and its associated lipotoxic metabolic effects. However, the development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter, we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition, and expandability capacity potential as well as molecular and metabolic characteristic signatures in both physiological and pathophysiological conditions. Current antilipotoxic strategies for future clinical application are also discussed in this chapter.
Collapse
Affiliation(s)
- Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| | - Vanessa Pellegrinelli
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| |
Collapse
|
47
|
Halasz L, Divoux A, Sandor K, Erdos E, Daniel B, Smith SR, Osborne TF. An Atlas of Promoter Chromatin Modifications and HiChIP Regulatory Interactions in Human Subcutaneous Adipose-Derived Stem Cells. Int J Mol Sci 2023; 25:437. [PMID: 38203607 PMCID: PMC10778978 DOI: 10.3390/ijms25010437] [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: 10/23/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
The genome of human adipose-derived stem cells (ADSCs) from abdominal and gluteofemoral adipose tissue depots are maintained in depot-specific stable epigenetic conformations that influence cell-autonomous gene expression patterns and drive unique depot-specific functions. The traditional approach to explore tissue-specific transcriptional regulation has been to correlate differential gene expression to the nearest-neighbor linear-distance regulatory region defined by associated chromatin features including open chromatin status, histone modifications, and DNA methylation. This has provided important information; nonetheless, the approach is limited because of the known organization of eukaryotic chromatin into a topologically constrained three-dimensional network. This network positions distal regulatory elements in spatial proximity with gene promoters which are not predictable based on linear genomic distance. In this work, we capture long-range chromatin interactions using HiChIP to identify remote genomic regions that influence the differential regulation of depot-specific genes in ADSCs isolated from different adipose depots. By integrating these data with RNA-seq results and histone modifications identified by ChIP-seq, we uncovered distal regulatory elements that influence depot-specific gene expression in ADSCs. Interestingly, a subset of the HiChIP-defined chromatin loops also provide previously unknown connections between waist-to-hip ratio GWAS variants with genes that are known to significantly influence ADSC differentiation and adipocyte function.
Collapse
Affiliation(s)
- Laszlo Halasz
- Division of Diabetes Endocrinology and Metabolism, Departments of Medicine, Biological Chemistry and Pediatrics, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA (T.F.O.)
| | - Adeline Divoux
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA;
| | - Katalin Sandor
- Division of Diabetes Endocrinology and Metabolism, Departments of Medicine, Biological Chemistry and Pediatrics, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA (T.F.O.)
| | - Edina Erdos
- Division of Diabetes Endocrinology and Metabolism, Departments of Medicine, Biological Chemistry and Pediatrics, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA (T.F.O.)
| | - Bence Daniel
- Division of Diabetes Endocrinology and Metabolism, Departments of Medicine, Biological Chemistry and Pediatrics, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA (T.F.O.)
| | - Steven R. Smith
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA;
| | - Timothy F. Osborne
- Division of Diabetes Endocrinology and Metabolism, Departments of Medicine, Biological Chemistry and Pediatrics, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA (T.F.O.)
| |
Collapse
|
48
|
Liu W, Liu T, Zhao Q, Ma J, Jiang J, Shi H. Adipose Tissue-Derived Extracellular Vesicles: A Promising Biomarker and Therapeutic Strategy for Metabolic Disorders. Stem Cells Int 2023; 2023:9517826. [PMID: 38169960 PMCID: PMC10761228 DOI: 10.1155/2023/9517826] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/23/2023] [Accepted: 11/25/2023] [Indexed: 01/05/2024] Open
Abstract
Adipose tissue plays an important role in systemic energy metabolism, and its dysfunction can lead to severe metabolic disorders. Various cells in adipose tissue communicate with each other to maintain metabolic homeostasis. Extracellular vesicles (EVs) are recognized as novel medium for remote intercellular communication by transferring various bioactive molecules from parental cells to distant target cells. Increasing evidence suggests that the endocrine functions of adipose tissue and even the metabolic homeostasis are largely affected by different cell-derived EVs, such as insulin signaling, lipolysis, and metabolically triggered inflammation regulations. Here, we provide an overview focused on the role of EVs released by different cell types of adipose tissue in metabolic diseases and their possible molecular mechanisms and highlight the potential applications of EVs as biomarkers and therapeutic targets. Moreover, the current EVs-based therapeutic strategies have also been discussed. This trial is registered with NCT05475418.
Collapse
Affiliation(s)
- Wenhui Liu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Tianyan Liu
- Center of Laboratory Medicine, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
| | - Qingyu Zhao
- Department of Nephrology, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
| | - Junqiu Ma
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
- Center of Laboratory Medicine, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
- Center of Laboratory Medicine, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
| | - Hui Shi
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou 215600, Jiangsu, China
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| |
Collapse
|
49
|
Han SM, Park ES, Park J, Nahmgoong H, Choi YH, Oh J, Yim KM, Lee WT, Lee YK, Jeon YG, Shin KC, Huh JY, Choi SH, Park J, Kim JK, Kim JB. Unique adipose tissue invariant natural killer T cell subpopulations control adipocyte turnover in mice. Nat Commun 2023; 14:8512. [PMID: 38129377 PMCID: PMC10739728 DOI: 10.1038/s41467-023-44181-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Adipose tissue invariant natural killer T (iNKT) cells are a crucial cell type for adipose tissue homeostasis in obese animals. However, heterogeneity of adipose iNKT cells and their function in adipocyte turnover are not thoroughly understood. Here, we investigate transcriptional heterogeneity in adipose iNKT cells and their hierarchy using single-cell RNA sequencing in lean and obese mice. We report that distinct subpopulations of adipose iNKT cells modulate adipose tissue homeostasis through adipocyte death and birth. We identify KLRG1+ iNKT cells as a unique iNKT cell subpopulation in adipose tissue. Adoptive transfer experiments showed that KLRG1+ iNKT cells are selectively generated within adipose tissue microenvironment and differentiate into a CX3CR1+ cytotoxic subpopulation in obese mice. In addition, CX3CR1+ iNKT cells specifically kill enlarged and inflamed adipocytes and recruit macrophages through CCL5. Furthermore, adipose iNKT17 cells have the potential to secrete AREG, and AREG is involved in stimulating adipose stem cell proliferation. Collectively, our data suggest that each adipose iNKT cell subpopulation plays key roles in the control of adipocyte turnover via interaction with adipocytes, adipose stem cells, and macrophages in adipose tissue.
Collapse
Affiliation(s)
- Sang Mun Han
- National Leading Researcher 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
| | - Eun Seo Park
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Jeu Park
- National Leading Researcher 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 Leading Researcher 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
| | - Yoon Ha Choi
- Department of Life Sciences, POSTECH, Pohang, 37673, Republic of Korea
| | - Jiyoung Oh
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Kyung Min Yim
- National Leading Researcher 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
| | - Won Taek Lee
- National Leading Researcher 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
| | - Yun Kyung Lee
- Internal Medicine, Seoul National University College of Medicine & Seoul National University Bundang Hospital, Seoul, 03080, Republic of Korea
| | - Yong Geun Jeon
- National Leading Researcher 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 Cheul Shin
- National Leading Researcher 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
| | - Jin Young Huh
- Department of Life Science, Sogang University, Seoul, 04107, Republic of Korea
| | - Sung Hee Choi
- Internal Medicine, Seoul National University College of Medicine & Seoul National University Bundang Hospital, Seoul, 03080, Republic of Korea
| | - Jiyoung Park
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Jong Kyoung Kim
- Department of Life Sciences, POSTECH, Pohang, 37673, Republic of Korea.
| | - Jae Bum Kim
- National Leading Researcher 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
|
50
|
Rosca R, Paduraru DN, Bolocan A, Musat F, Ion D, Andronic O. A Comprehensive Review of Inguinal Hernia Occurrence in Obese Individuals. MAEDICA 2023; 18:692-698. [PMID: 38348082 PMCID: PMC10859212 DOI: 10.26574/maedica.2023.18.4.692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Inguinal hernia repair is one of the most commonly performed surgical activities worldwide. Given the circumstances, understanding and identifying the risk and the protective factors is an essential step in order to prevent, diagnose and treat such a common condition. For a long time, obesity was generally considered to be a risk factor in the occurrence of an inguinal hernia. Studies have provided some unexpected data, suggesting that it might actually be a protective factor. This review aims to provide an overview on this topic, taking into account systemic aspects such as collagen distribution and metabolism. In inguinal hernia patients, the ratio between type I collagen and type III collagen is decreased, with type III collagen being responsible for the weakness of the abdominal wall. In obese patients, the extracellular matrix becomes richer in collagen, especially type I collagen, which will generate strength and stiffness. Obesity seems to be a protective factor indeed, but in order to understand the underlying mechanism and to choose the optimal surgical approach, further research is needed.
Collapse
Affiliation(s)
- Ruxandra Rosca
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Dan Nicolae Paduraru
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- University Emergency Hospital Bucharest, Romania
| | - Alexandra Bolocan
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- University Emergency Hospital Bucharest, Romania
| | - Florentina Musat
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- University Emergency Hospital Bucharest, Romania
| | - Daniel Ion
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- University Emergency Hospital Bucharest, Romania
| | - Octavian Andronic
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- University Emergency Hospital Bucharest, Romania
| |
Collapse
|