1
|
Herz CT, Kulterer OC, Prager M, Marculescu R, Prager G, Kautzky-Willer A, Hacker M, Trajanoski S, Köfeler HC, Gallé B, Haug AR, Berry D, Kiefer FW. Bariatric surgery promotes recruitment of brown fat linked to alterations in the gut microbiota. Eur J Endocrinol 2025; 192:603-611. [PMID: 40366070 DOI: 10.1093/ejendo/lvaf081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/10/2025] [Indexed: 05/15/2025]
Abstract
OBJECTIVE The mechanisms of bariatric surgery-induced weight loss and metabolic improvements are still incompletely understood and reach beyond malabsorption or calorie restriction. We sought to investigate the effect of bariatric surgery on brown adipose tissue (BAT) activity and a potential connection with changes in energy metabolism, the gut microbiota, and short-chain fatty acid (SCFA) composition. METHODS We included 32 subjects (25 females) with morbid obesity and analyzed their metabolic profile, gut microbiota composition, circulating SCFAs, energy expenditure, and cold-induced BAT activity using [18F]Fluorodeoxyglucose-positron emission tomography-computed tomography before and up to 1 year after bariatric surgery. RESULTS Twelve months after surgery, the percentage of individuals with active BAT had increased from 28% to 53%. The BAT-negative (BATneg) individuals who had an adverse metabolic profile at baseline compared with subjects with active BAT (BATpos) showed a greater metabolic benefit after surgery. While no changes in overall gut bacterial diversity were observed between BATpos and BATneg, the abundance of 3 specific bacterial families, including Akkermansiaceae, Pasteurellaceae, and Carnobacteriaceae, was distinctly regulated between BAT groups. The bacterial genera most strongly increased in BATpos vs BATneg subjects were all positively correlated with BAT volume and BAT activity. Finally, circulating concentrations of the SCFAs acetate, butyrate, and propionate rose after bariatric surgery and were related to bacterial genera such as Akkermansia, Dialister, and Lachnospiraceae FCS020 group, all known SCFA producers. CONCLUSIONS Bariatric surgery helps recruit active BAT in individuals with obesity and is linked to distinct alterations in the gut microbiome and SCFA composition. TRIAL REGISTRATION NUMBER ClinicalTrials.gov (NCT03168009).
Collapse
Affiliation(s)
- Carsten T Herz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Oana C Kulterer
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Marlene Prager
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Rodrig Marculescu
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Gerhard Prager
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Alexandra Kautzky-Willer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Harald C Köfeler
- Core Facility Mass Spectrometry, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Birgit Gallé
- Core Facility Molecular Biology, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Alexander R Haug
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - David Berry
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Florian W Kiefer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Zhang J, Shen M, Yin Y, Chen Y, Deng X, Mo J, Zhou X, Lin J, Chen X, Xie X, Wu X, Chen X. Carnosic acid reduces lipid content, enhances gut health, and modulates microbiota composition and metabolism in diet-induced obese mice. Food Funct 2025; 16:1888-1902. [PMID: 39932492 DOI: 10.1039/d4fo04534c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Carnosic acid (CA) is a bioactive phenolic diterperne compound found in sage and rosemary. The present study investigated the beneficial effects of CA (50 and 100 mg per kg bw) in diet-induced obese mice and the underlying mechanisms of action. After the intervention, the physiology, lipid metabolism, and tissue morphology, as well as the inflammation, gut microbiota, and metabolomics in the colon were measured. We found that CA improved the composition and metabolism of the gut microbiota in obese mice, with Akkermansia being the dominant bacterium negatively correlated with obesity and various fecal metabolites. Regarding the intestinal barrier function, CA promoted the expression of tight junction proteins and inhibited the TLR4/MyD88/NF-κB signaling pathway in obese mice to alleviate colonic inflammation. These results suggest that CA improved multiple aspects of gut health in diet-induced obesity in mice, providing a scientific basis for future clinical studies in humans.
Collapse
Affiliation(s)
- Jing Zhang
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Mengzhu Shen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yue Yin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yuru Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xianying Deng
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Jingyun Mo
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xiaoling Zhou
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Juanying Lin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinxin Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinwei Xie
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xian Wu
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, Ohio 45056, USA.
| | - Xuexiang Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| |
Collapse
|
3
|
Yu J, Gu X, Guo Y, Gao M, Cheng S, Meng M, Cui X, Zhang Z, Guo W, Yan D, Sheng M, Zhai L, Ji J, Ma X, Li Y, Cao Y, Wu X, Zhao J, Hu Y, Tan M, Lu Y, Xu L, Liu B, Hu C, Ma X. E3 ligase FBXW7 suppresses brown fat expansion and browning of white fat. EMBO Rep 2025; 26:748-767. [PMID: 39747664 PMCID: PMC11811183 DOI: 10.1038/s44319-024-00337-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] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/26/2024] [Accepted: 11/08/2024] [Indexed: 01/04/2025] Open
Abstract
Thermogenic fat, including brown and beige fat, dissipates heat via thermogenesis and enhances energy expenditure. Thus, its activation represents a therapeutic strategy to combat obesity. Here, we demonstrate that levels of F-box and WD repeat domain-containing 7 (FBXW7), an E3 ubiquitin protein ligase, negatively correlate with thermogenic fat functionality. FBXW7 overexpression in fat suppresses energy expenditure and thermogenesis, thus aggravates obesity and metabolic dysfunctions in mice. Conversely, FBXW7 depletion in fat leads to brown fat expansion and browning of white fat, and protects mice from diet induced obesity, hepatic steatosis, and hyperlipidemia. Mechanistically, FBXW7 binds to S6K1 and promotes its ubiquitination and proteasomal degradation, which in turn impacts glycolysis and brown preadipocyte proliferation via lactate. Besides, the beneficial metabolic effects of FBXW7 depletion in fat are attenuated by fat-specific knockdown of S6K1 in vivo. In summary, we provide evidence that adipose FBXW7 acts as a major regulator for thermogenic fat biology and energy homeostasis and serves as potential therapeutic target for obesity and metabolic diseases.
Collapse
Grants
- 32325024,82300979,32222024,32271224,32071148,22225702,82000802 MOST | National Natural Science Foundation of China (NSFC)
- 2023YFA1800400,2019YFA09004500 MOST | National Key Research and Development Program of China (NKPs)
- 22ZR1421200,21140904300 Science and Technology Commission of Shanghai Municipality (STCSM)
- CSTB2022NSCQ-JQX0033 Natural Science Foundation of Chongqing, China
- 2021C03069 Key Research and Development Project of Zhejiang Province, China
- LY20H070003 Zhejiang Provincial Natural Science Foundation of China
- SHSMU-ZDCX20212700 Innovation research team of high-level local universities in Shanghai
- 2022ZZ01002 Shanghai Research Center for Endocrine and Metabolic Diseases
- 2023M741184 China Postdoctoral Science Foundation(China Postdoctoral Foundation Project)
Collapse
Affiliation(s)
- Jian Yu
- Joint Center for Translational Medicine, Fengxian District Central Hospital, Fengxian District, Shanghai, 201400, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xuejiang Gu
- Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yingying Guo
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai, 200233, China
| | - Mingyuan Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Shimiao Cheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Meiyao Meng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiangdi Cui
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhe Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wenxiu Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Dandan Yan
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai, 200233, China
| | - Maozheng Sheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Linhui Zhai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jing Ji
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xinhui Ma
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yu Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yuxiang Cao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xia Wu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiejie Zhao
- Ministry of Education Key Laboratory of Metabolism and Molecular Medicine, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200000, China
| | - Yepeng Hu
- Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yan Lu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai, 200233, China
- Ministry of Education Key Laboratory of Metabolism and Molecular Medicine, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200000, China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Institute for Aging, East China Normal University, Shanghai, 200241, China.
| | - Bin Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Cheng Hu
- Joint Center for Translational Medicine, Fengxian District Central Hospital, Fengxian District, Shanghai, 201400, China.
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai, 200233, China.
| | - Xinran Ma
- Joint Center for Translational Medicine, Fengxian District Central Hospital, Fengxian District, Shanghai, 201400, China.
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China.
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Institute for Aging, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
4
|
De Francesco F, Sbarbati A, Sierra LAQ, Zingaretti N, Sarmadian Z, Parodi PC, Ricci G, Riccio M, Mobasheri A. Anatomy, Histology, and Embryonic Origin of Adipose Tissue: Insights to Understand Adipose Tissue Homofunctionality in Regeneration and Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1474:53-78. [PMID: 39107527 DOI: 10.1007/5584_2024_801] [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: 01/05/2025]
Abstract
Preadipocytes are formed during the 14th and 16th weeks of gestation. White adipose tissue, in particular, is generated in specific areas and thereby assembles after birth, rapidly increasing following the propagation of adipoblasts, which are considered the preadipocyte cell precursors. The second trimester of gestation is a fundamental phase of adipogenesis, and in the third trimester, adipocytes, albeit small may be present within the main deposition areas. In the course of late gestation, adipose tissue develops in the foetus and promotes the synthesis of large amounts of uncoupling protein 1, in similar quantities relative to differentiated brown adipose tissue. In mammals, differentiation occurs in two functionally different types of adipose cells: white adipose cells resulting from lipid storage and brown adipose cells from increased metabolic energy consumption. During skeletogenesis, synovial joints develop through the condensation of mesenchymal cells, which forms an insertional layer of flattened cells that umlaut skeletal elements, by sharing the same origin in the development of synovium. Peri-articular fat pads possess structural similarity with body subcutaneous white adipose tissue; however, they exhibit a distinct metabolic function due to the micro-environmental cues in which they are embedded. Fat pads are an important component of the synovial joint and play a key role in the maintenance of joint homeostasis. They are also implicated in pathological states such as osteoarthritis.In this paper we explore the therapeutic potential of adipocyte tissue mesenchymal precursor-based stem cell therapy linking it back to the anatomic origin of adipose tissue.
Collapse
Affiliation(s)
- Francesco De Francesco
- Department of Reconstructive Surgery and Hand Surgery, AOU Ospedali Riuniti delle Marche, Ancona, Italy
| | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, Verona, Italy
| | | | - Nicola Zingaretti
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, Udine, Italy
| | - Zahra Sarmadian
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Pier Camillo Parodi
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, Udine, Italy
| | - Giulia Ricci
- Department of Experimental Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Michele Riccio
- Department of Reconstructive Surgery and Hand Surgery, AOU Ospedali Riuniti delle Marche, Ancona, Italy
| | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
- Department of Joint Surgery, Sun Yat-sen University, Guangzhou, People's Republic of China.
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium.
| |
Collapse
|
5
|
Bahadoran Z, Mirmiran P, Kashfi K, Ghasemi A. Effects of time-restricted feeding (TRF)-model of intermittent fasting on adipose organ: a narrative review. Eat Weight Disord 2024; 29:77. [PMID: 39719521 DOI: 10.1007/s40519-024-01709-w] [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: 06/19/2024] [Accepted: 12/11/2024] [Indexed: 12/26/2024] Open
Abstract
Time-restricted feeding (TRF), an intermittent fasting approach involving a shortened eating window within 24 h, has gained popularity as a weight management approach. This review addresses how TRF may favor fat redistribution and the function of the adipose organ. TRF trials (mainly 16:8 model, with a duration of 5-48 weeks) reported a significant weight loss (1.2-10.2%, ~ 1.4-9.4 kg), with a considerable decrease in total fat mass (1.6-21%, ~ 0.5-7 kg) and visceral adipose compartment (VAC, 11-27%) in overweight and obese subjects. Experimental TRF in normal-fed and obesogenic-diet-fed mice and rats (with a fasting duration ranging between 9 and 21 h within 1-17 weeks) reported a significant reduction in body weight (~ 7-40%), total fat mass (~ 17-71%), and intrahepatic fat (~ 25-72%). TRF also improves VAC and subcutaneous adipose compartment (SAC) function by decreasing adipocyte size, macrophage infiltration, M1-macrophage polarity, and downregulating inflammatory genes. In conclusion, beyond its effect on body weight loss, total fat mass, and intrahepatic fat accumulation, TRF favors adipose organ fat redistribution in overweight and obese subjects by decreasing VAC and improving the function of VAC and SAC.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Micronutrient Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Sahid-Erabi St, Yemen St, Chamran Exp, Tehran, Iran.
| |
Collapse
|
6
|
Morciano C, Gugliandolo S, Capece U, Di Giuseppe G, Mezza T, Ciccarelli G, Soldovieri L, Brunetti M, Avolio A, Splendore A, Pontecorvi A, Giaccari A, Cinti F. SGLT2 inhibition and adipose tissue metabolism: current outlook and perspectives. Cardiovasc Diabetol 2024; 23:449. [PMID: 39702365 PMCID: PMC11660748 DOI: 10.1186/s12933-024-02539-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
Sodium-glucose co-transporter 2 inhibitors (SGLT2i) have emerged as important agents for the treatment of type 2 diabetes mellitus (T2DM). SGLT2 inhibitors have been associated with improved cardiovascular outcomes, not only through their immediate hemodynamic effects-such as glycosuria and (at least temporary) increased natriuresis-but also due to their multifaceted impact on metabolism. Recently, studies have also focused on the effects of SGLT2 inhibitors on adipose tissue. Aside from the well-documented effects on human adiposity, SGLT2i have shown, both in vitro and in murine models, the ability to reduce fat mass, upregulate genes related to browning of white adipose tissue, influence adipocyte size and fatty acid oxidation, and improve oxidative stress and overall metabolic health. In humans, even though data are still limited, recent evidence seems to confirm that the SGLT2i effects observed in cardiovascular outcome trials could be partially explained by their impact on adipose tissue. This review aims to clarify the impact of SGLT2i on adipose tissue, highlighting their role in metabolic health and their potential to transform treatment strategies for T2DM beyond glucose metabolism.
Collapse
Affiliation(s)
- Cassandra Morciano
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Scienze Cliniche e Sperimentali, Medicina Interna - Università degli studi di Brescia, Brescia, BS, Italy
| | - Shawn Gugliandolo
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Umberto Capece
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gianfranco Di Giuseppe
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Teresa Mezza
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Pancreas Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Gea Ciccarelli
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Laura Soldovieri
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Michela Brunetti
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Adriana Avolio
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Amelia Splendore
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alfredo Pontecorvi
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Giaccari
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Francesca Cinti
- Centro Malattie Endocrine e Metaboliche, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
7
|
Grigorova N, Ivanova Z, Petrova V, Vachkova E, Beev G. Supernatants from Newly Isolated Lacticaseibacillus paracasei P4 Ameliorate Adipocyte Metabolism in Differentiated 3T3-L1 Cells. Biomedicines 2024; 12:2785. [PMID: 39767692 PMCID: PMC11673354 DOI: 10.3390/biomedicines12122785] [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: 10/16/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Background:Lacticaseibacillus paracasei (L. paracasei) strains and their postbiotics show potential for managing metabolic disorders such as diabetes and obesity. Two newly isolated L. paracasei strains, M2.1 and P4, were yielded from Formica rufa anthills in Sinite Kamani National Park, Bulgaria. Their metabolic effects on mature 3T3-L1 adipocytes were investigated. Methods: Mature 3T3-L1 adipocytes were treated for 24 h with 10% (v/v) cell-free supernatants (CFSs) of M2.1 or P4. Two experimental (M2.1, P4) and two control groups (mature, untreated adipocytes and mature adipocytes, treated with 10% (v/v) MRS broth) were analyzed for intracellular lipid accumulation, glucose uptake, and the mRNA expression of lipid metabolism and beta-oxidation-related genes. Fold changes in gene expression were assessed using RT-qPCR. Results: Both M2.1 and P4 CFSs enhanced glucose uptake by over 30% compared to the control. P4 demonstrated a more favorable effect by significantly upregulating adipose triglyceride lipase-patatin-like phospholipase domain containing 2, adiponectin, and peroxisomal beta-oxidation enzymes-acyl-coenzyme A oxidase 1, palmitoyl. Intracellular lipid accumulation increased only with M2.1, while P4 supported improved lipid turnover without promoting excessive lipid storage or lipolysis. Conclusions: P4 CFS exhibits the potential to improve adipocyte metabolism by enhancing glucose uptake, promoting beta-oxidation, and increasing adiponectin expression, offering a promising strategy for managing metabolic dysfunctions.
Collapse
Affiliation(s)
- Natalia Grigorova
- Department of Pharmacology, Animal Physiology, Biochemistry and Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria; (N.G.); (Z.I.); (V.P.); (E.V.)
| | - Zhenya Ivanova
- Department of Pharmacology, Animal Physiology, Biochemistry and Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria; (N.G.); (Z.I.); (V.P.); (E.V.)
| | - Valeria Petrova
- Department of Pharmacology, Animal Physiology, Biochemistry and Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria; (N.G.); (Z.I.); (V.P.); (E.V.)
| | - Ekaterina Vachkova
- Department of Pharmacology, Animal Physiology, Biochemistry and Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria; (N.G.); (Z.I.); (V.P.); (E.V.)
| | - Georgi Beev
- Department of Biochemistry, Microbiology and Physics, Faculty of Agriculture, Trakia University, 6000 Stara Zagora, Bulgaria
| |
Collapse
|
8
|
Kuhnert LRB, Pontes RDFC, Neto JGO, Romão JS, Pinto CEDC, Oliveira KJ. Cinnamon powder intake enhances the effect of caloric restriction on white adipose tissue in male rats. J Mol Histol 2024; 56:19. [PMID: 39627596 DOI: 10.1007/s10735-024-10288-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/30/2024] [Indexed: 02/07/2025]
Abstract
Caloric Restriction (CR) and cinnamon promote several benefits, including the modulation of lipid metabolism and body fat mass. We hypothesize that cinnamon may act as a mimetic of restriction or enhance the effects of caloric restriction on adipose tissue. Adult male Wistar rats were divided into Control (CT, n = 8) and Cinnamon (CIN, n = 7), with free access to standard chow; Calorie Restriction (CR, n = 8) and Calorie Restriction with Cinnamon (CIN-CR, n = 7), subjected to a 30% reduction in food intake compared to the average consumption of CT rats. Both CIN groups received 50 mg cinnamon powder (Cinnamomun verum) per kg body mass, by gavage, over 6 weeks. Cinnamon treatment did not alter food intake under either ad libitum or caloric restriction conditions. The CR and CIN-CR groups exhibit lower body mass. Basal glycemia, lipid profile, and triglyceride-glycemic index were similar between groups. The combination of both interventions induced lower visceral white adipose tissue (WAT) mass, and smaller adipocyte diameter in the visceral and subcutaneous WAT compartments, accompanied by reduced expression of genes related to lipid metabolism (Acaca, Fasn, Cd36, Srebf1c), suggesting decreased lipid synthesis. Histological analyses identified a browning phenotype in the CR, CIN, and CIN-CR groups, positive for UCP1 immunostaining. The CR and CIN-CR groups showed lower Atg7 expression, and CIN-CR animals expressed increased levels of Lamp2, suggesting modulation of autophagy. Brown adipose tissue mass and lipid content were not influenced by any intervention. These findings suggest that cinnamon may enhance the effects of caloric restriction in promoting adipocyte metabolic health.
Collapse
Affiliation(s)
- Lia Rafaella Ballard Kuhnert
- Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
- Laboratory of Experimental Pathology, Department of Immunobiology, Institute of Biology, Federal Fluminense University, Niteroi, RJ, Brazil
| | | | - Jessika Geisebel Oliveira Neto
- Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Juliana Santos Romão
- Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Carla Eponina de Carvalho Pinto
- Laboratory of Experimental Pathology, Department of Immunobiology, Institute of Biology, Federal Fluminense University, Niteroi, RJ, Brazil
| | - Karen Jesus Oliveira
- Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil.
| |
Collapse
|
9
|
Zheng S, Tan Y, Yang S, Quan Z. Evaluation Between Serum Concentrations of Lipocalin-2 and Metabolic Syndrome and its Components in Korean-Chinese and Han-Chinese Individuals from Yanbian Area. Metab Syndr Relat Disord 2024; 22:735-742. [PMID: 39029476 DOI: 10.1089/met.2024.0099] [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: 07/21/2024] Open
Abstract
Objectives: To investigate the association between the blood concentration of lipocalin-2 (LCN2) in local multiethnic residents and the increased risk for the development of metabolic syndrome (MS) in the Yanbian Korean Autonomous Prefecture population. Methods: A total of 2078 subjects with (study group) or without (control group) MS (1217 Korean-Chinese and 861 Han-Chinese subjects) were included in this study. MS subjects were divided into five groups according to ethnicity and MS components. They were assessed for smoking history, drinking history, past medical history, general demographic characteristics, and LCN2 concentrations. Results: LCN2 concentrations were higher in all ethnic MS groups than in the control group, and the highest concentrations were detected in Han-Chinese subjects with dyslipidemia. Moreover, LCN2 concentrations were significantly higher in Korean-Chinese individuals with all MS components than in the control group. Logistic regression analyses were conducted. In the unadjusted models, Korean-Chinese and Han-Chinese individuals with high LCN2 concentrations both faced a risk of MS with odds ratios (ORs) of 2.339 (95% confidence interval [CI]: 1.632-3.352) and 1.523 (95% CI: 1.101-2. 108), respectively. After the adjustment, the risk only remained in Korean-Chinese individuals, with an OR of 1.818 (95% CI: 1.031-3.207). Conclusion: Elevated circulating LCN2 was associated with the increased incidence of MS, and the effect in Korean-Chinese individuals was stronger than that in Han-Chinese individuals.
Collapse
Affiliation(s)
- Songyun Zheng
- Department of Clinical Medicine, Medical College, Yanbian University, Yanji City, China
| | - Yuanyuan Tan
- Department of Clinical Medicine, Medical College, Yanbian University, Yanji City, China
| | - Shuhan Yang
- Department of Clinical Medicine, Medical College, Yanbian University, Yanji City, China
| | - Zhenyu Quan
- Department of Preventive Medicine, Medical College, Yanbian University, Yanji City, China
| |
Collapse
|
10
|
Hosain O, Clinkenbeard EL. Adiposity and Mineral Balance in Chronic Kidney Disease. Curr Osteoporos Rep 2024; 22:561-575. [PMID: 39394545 DOI: 10.1007/s11914-024-00884-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 10/13/2024]
Abstract
PURPOSE OF REVIEW Bone homeostasis is balanced between formation and resorption activities and remain in relative equilibrium. Under disease states this process is disrupted, favoring more resorption over formation, leading to significant bone loss and fracture incidence. This aspect is a hallmark for patients with chronic kidney disease mineral and bone disorder (CKD-MBD) affecting a significant portion of the population, both in the United States and worldwide. Further study into the underlying effects of the uremic microenvironment within bone during CKD-MBD are critical as fracture incidence in this patient population not only leads to increased morbidity, but also increased mortality. Lack of bone homeostasis also leads to mineral imbalance contributing to cardiovascular calcifications. One area understudied is the possible involvement of bone marrow adipose tissue (BMAT) during the progression of CKD-MBD. RECENT FINDINGS BMAT accumulation is found during aging and in several disease states, some of which overlap as CKD etiologies. Importantly, research has found presence of BMAT inversely correlates with bone density and volume. Understanding the underlying molecular mechanisms for BMAT formation and accumulation during CKD-MBD may offer a potential therapeutic avenue to improve bone homeostasis and ultimately mineral metabolism.
Collapse
Affiliation(s)
- Ozair Hosain
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN, 46022, USA
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Erica L Clinkenbeard
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
11
|
Shaikh I, Bhatt LK. Targeting Adipokines: A Promising Therapeutic Strategy for Epilepsy. Neurochem Res 2024; 49:2973-2987. [PMID: 39060767 DOI: 10.1007/s11064-024-04219-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Epilepsy affects 65 million people globally and causes neurobehavioral, cognitive, and psychological defects. Although research on the disease is progressing and a wide range of treatments are available, approximately 30% of people have refractory epilepsy that cannot be managed with conventional medications. This underlines the importance of further understanding the condition and exploring cutting-edge targets for treatment. Adipokines are peptides secreted by adipocyte's white adipose tissue, involved in controlling food intake and metabolism. Their regulatory functions in the central nervous system (CNS) are multifaceted and identified in several physiology and pathologies. Adipokines play a role in oxidative stress and neuroinflammation which are associated with brain degeneration and connected neurological diseases. This review aims to highlight the potential impacts of leptin, adiponectin, apelin, vaspin, visfatin, and chimerin in the pathogenesis of epilepsy.
Collapse
Affiliation(s)
- Iqraa Shaikh
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
12
|
Turyn J, Stelmanska E, Szrok-Jurga S. Two Regions with Different Expression of Lipogenic Enzymes in Rats' Posterior Subcutaneous Fat Depot. Int J Mol Sci 2024; 25:11546. [PMID: 39519099 PMCID: PMC11546078 DOI: 10.3390/ijms252111546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/20/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Lipid metabolism in various adipose tissue depots can differ vastly. This also applies to lipogenesis, the process of synthesizing fatty acids from acetyl-CoA. This study compared the expression of some lipogenic enzymes: fatty acid synthase (FASN), ATP-citrate lyase (ACLY), and malic enzyme 1 (ME1) in different regions of the posterior subcutaneous adipose tissue in rats. Methods and Results: Posterior subcutaneous adipose tissue collected from twelve-month-old Wistar rats was divided into six parts (A-F). The expression of genes encoding lipogenic enzymes was assessed by measuring their activity and mRNA levels using real-time PCR. In the gluteal region of the fat pad, there were much higher levels of activity and mRNA for these lipogenic enzymes compared to the dorsolumbar region. The mRNA level of FASN increased by more than twentyfold, whereas the level of ME1 and ACLY increased eight- and fivefold respectively. This phenomenon was observed in both old and young animals. Furthermore, the lack of uncoupling protein one (Ucp1) expression suggests that neither the presence of brown adipocytes in the gluteal part nor the transformation of white adipocytes into beige contributed to the observed differences. Conclusion: These results indicate that the gluteal white adipose tissue appears to be a unique and separate subcutaneous fat depot.
Collapse
Affiliation(s)
- Jacek Turyn
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | | | - Sylwia Szrok-Jurga
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland;
| |
Collapse
|
13
|
Valladolid-Acebes I. Hippocampal Leptin Resistance and Cognitive Decline: Mechanisms, Therapeutic Strategies and Clinical Implications. Biomedicines 2024; 12:2422. [PMID: 39594988 PMCID: PMC11591892 DOI: 10.3390/biomedicines12112422] [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: 09/13/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Leptin, an adipokine essential for regulating energy balance, exerts important effects on brain function, notably within the hippocampus, a region integral to learning and memory. Leptin resistance, characterized by diminished responsiveness to elevated leptin levels, disrupts hippocampal function and exacerbates both obesity and cognitive impairments. Scope: This review critically examines how leptin resistance impairs hippocampal synaptic plasticity processes, specifically affecting long-term potentiation (LTP) and long-term depression (LTD), which are crucial for cognitive performance. Findings: Recent research highlights that leptin resistance disrupts N-methyl-D-aspartate (NMDA) receptor dynamics and hippocampal structure, leading to deficits in spatial learning and memory. Additionally, high-fat diets (HFDs), which contribute to leptin resistance, further deteriorate hippocampal function. Potential therapeutic strategies, including leptin sensitizers, show promise in mitigating brain disorders associated with leptin resistance. Complementary interventions such as caloric restriction and physical exercise also enhance leptin sensitivity and offer potential benefits to alleviating cognitive impairments. Aims of the review: This review synthesizes recent findings on the molecular pathways underlying leptin resistance and its impact on synaptic transmission and plasticity in the hippocampus. By identifying potential therapeutic targets, this work aims to provide an integrated approach for addressing cognitive deficits in obesity, ultimately improving the quality of life for affected individuals.
Collapse
Affiliation(s)
- Ismael Valladolid-Acebes
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| |
Collapse
|
14
|
Wang Z, Zhou C, Meng L, Mo X, Xie D, Huang X, He X, Luo S, Qin H, Li Q, Lai S. Development and validation of an MRI and clinicopathological factors prediction model for low anterior resection syndrome in anterior resection of middle and low rectal cancer. Heliyon 2024; 10:e36498. [PMID: 39296093 PMCID: PMC11409036 DOI: 10.1016/j.heliyon.2024.e36498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/21/2024] Open
Abstract
OBJECTIVE To validate the predictive power of newly developed magnetic resonance (MR) morphological and clinicopathological risk models in predicting low anterior resection syndrome (LARS) 6 months after anterior resection of middle and low rectal cancer (MLRC). METHODS From May 2018 to January 2021, 236 patients with MLRC admitted to two hospitals (internal and external validation) were included. MR images, clinicopathological data, and LARS scores (LARSS) were collected. Tumor morphology data included longitudinal involvement length, maximum tumor diameter, proportion of tumor to circumference of the intestinal wall, tumor mesorectal infiltration depth, circumferential margin status, and distance between the tumor and anal margins. Pelvic measurements included anorectal angle, mesenterial volume (MRV), and pelvic volume. Univariate and multivariate logistic regression was used to obtain independent risk factors of LARS after anterior resection Then, the prediction model was constructed, expressed as a nomogram, and its internal and external validity was assessed using receiver operating characteristic curves. RESULTS The uni- and multivariate analysis revealed distance between the tumor and anal margins, MRV, pelvic volume, and body weight as significant independent risk factors for predicting LARS. From the nomogram, the area under the curve (AUC), sensitivity, and specificity were 0.835, 75.0 %, and 80.4 %, respectively. The AUC, sensitivity, and specificity in the external validation group were 0.874, 83.3 %, and 91.7 %, respectively. CONCLUSION This study shows that MR imaging and clinicopathology presented by a nomogram can strongly predict LARSS, which can then individually predict LARS 6 months after anterior resection in patients with MLRC and facilitate clinical decision-making. CLINICAL RELEVANCE STATEMENT We believe that our study makes a significant contribution to the literature. This method of predicting postoperative anorectal function by preoperative measurement of MRV provides a new tool for clinicians to study LARS.
Collapse
Affiliation(s)
- Zheng Wang
- Medical Imaging Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Chuanji Zhou
- Department of Radiology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Linghou Meng
- Department of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Xianwei Mo
- Department of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Dong Xie
- Medical Imaging Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Xiaoliang Huang
- Department of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Xinxin He
- Department of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Shanshan Luo
- Department of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Haiquan Qin
- Department of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Qiang Li
- Medical Imaging Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Shaolv Lai
- Medical Imaging Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| |
Collapse
|
15
|
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: 2] [Impact Index Per Article: 2.0] [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
|
16
|
Röszer T. MicroRNA Profile of Mouse Adipocyte-Derived Extracellular Vesicles. Cells 2024; 13:1298. [PMID: 39120327 PMCID: PMC11311276 DOI: 10.3390/cells13151298] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
The post-transcriptional control of gene expression is a complex and evolving field in adipocyte biology, with the premise that the delivery of microRNA (miRNA) species to the obese adipose tissue may facilitate weight loss. Cells shed extracellular vesicles (EVs) that may deliver miRNAs as intercellular messengers. However, we know little about the miRNA profile of EVs secreted by adipocytes during postnatal development. Here, we defined the miRNA cargo of EVs secreted by mouse adipocytes in two distinct phases of development: on postnatal day 6, when adipocytes are lipolytic and thermogenic, and on postnatal day 56, when adipocytes have active lipogenesis. EVs were collected from cell culture supernatants, and their miRNA profile was defined by small RNA sequencing. The most abundant miRNA of mouse adipocyte-derived EVs was mmu-miR-148a-3p. Adipocyte EVs on postnatal day 6 were hallmarked with mmu-miR-98-5p, and some miRNAs were specific to this developmental stage, such as mmu-miR-466i-5p and 12 novel miRNAs. Adipocytes on postnatal day 56 secreted mmu-miR-365-3p, and 16 miRNAs were specific to this developmental stage. The miRNA cargo of adipocyte EVs targeted gene networks of cell proliferation, insulin signaling, interferon response, thermogenesis, and lipogenesis. We provided here a database of miRNAs secreted by developing mouse adipocytes, which may be a tool for further studies on the regulation of gene networks that control mouse adipocyte development.
Collapse
Affiliation(s)
- Tamás Röszer
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
17
|
Chen J, Pan Y, Lu Y, Fang X, Ma T, Chen X, Wang Y, Fang X, Zhang C, Song C. The Function and Mechanism of Long Noncoding RNAs in Adipogenic Differentiation. Genes (Basel) 2024; 15:875. [PMID: 39062654 PMCID: PMC11275360 DOI: 10.3390/genes15070875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/16/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Adipocytes are crucial for maintaining energy balance. Adipocyte differentiation involves distinct stages, including the orientation stage, clone amplification stage, clone amplification termination stage, and terminal differentiation stage. Understanding the regulatory mechanisms governing adipogenic differentiation is essential for comprehending the physiological processes and identifying potential biomarkers and therapeutic targets for metabolic diseases, ultimately improving glucose and fat metabolism. Adipogenic differentiation is influenced not only by key factors such as hormones, the peroxisome proliferator-activated receptor (PPAR) family, and the CCATT enhancer-binding protein (C/EBP) family but also by noncoding RNA, including microRNA (miRNA), long noncoding RNA (lncRNA), and circular RNA (circRNA). Among these, lncRNA has been identified as a significant regulator in adipogenic differentiation. Research has demonstrated various ways in which lncRNAs contribute to the molecular mechanisms of adipogenic differentiation. Throughout the adipogenesis process, lncRNAs modulate adipocyte differentiation and development by influencing relevant signaling pathways and transcription factors. This review provides a brief overview of the function and mechanism of lncRNAs in adipogenic differentiation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Chunlei Zhang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (J.C.); (Y.P.); (Y.L.); (X.F.); (T.M.); (X.C.); (Y.W.); (X.F.)
| | - Chengchuang Song
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (J.C.); (Y.P.); (Y.L.); (X.F.); (T.M.); (X.C.); (Y.W.); (X.F.)
| |
Collapse
|
18
|
Proença AB, Medeiros GR, Reis GDS, Losito LDF, Ferraz LM, Bargut TCL, Soares NP, Alexandre-Santos B, Campagnole-Santos MJ, Magliano DC, Nobrega ACLD, Santos RAS, Frantz EDC. Adipose tissue plasticity mediated by the counterregulatory axis of the renin-angiotensin system: Role of Mas and MrgD receptors. J Cell Physiol 2024; 239:e31265. [PMID: 38577921 DOI: 10.1002/jcp.31265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024]
Abstract
The renin-angiotensin system (RAS) is an endocrine system composed of two main axes: the classical and the counterregulatory, very often displaying opposing effects. The classical axis, primarily mediated by angiotensin receptors type 1 (AT1R), is linked to obesity-associated metabolic effects. On the other hand, the counterregulatory axis appears to exert antiobesity effects through the activation of two receptors, the G protein-coupled receptor (MasR) and Mas-related receptor type D (MrgD). The local RAS in adipose organ has prompted extensive research into white adipose tissue and brown adipose tissue (BAT), with a key role in regulating the cellular and metabolic plasticity of these tissues. The MasR activation favors the brown plasticity signature in the adipose organ by improve the thermogenesis, adipogenesis, and lipolysis, decrease the inflammatory state, and overall energy homeostasis. The MrgD metabolic effects are related to the maintenance of BAT functionality, but the signaling remains unexplored. This review provides a summary of RAS counterregulatory actions triggered by Mas and MrgD receptors on adipose tissue plasticity. Focus on the effects related to the morphology and function of adipose tissue, especially from animal studies, will be given targeting new avenues for treatment of obesity-associated metabolic effects.
Collapse
Affiliation(s)
- Ana Beatriz Proença
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Gabriela Rodrigues Medeiros
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Guilherme Dos Santos Reis
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Luiza da França Losito
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Luiza Mazzali Ferraz
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Thereza Cristina Lonzetti Bargut
- Department of Basic Sciences, Nova Friburgo Health Institute, Fluminense Federal University, Nova Friburgo, Rio de Janeiro, Brazil
| | - Nícia Pedreira Soares
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Beatriz Alexandre-Santos
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Maria Jose Campagnole-Santos
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - D'Angelo Carlo Magliano
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Antonio Claudio Lucas da Nobrega
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Robson Augusto Souza Santos
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Eliete Dalla Corte Frantz
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| |
Collapse
|
19
|
Perugini J, Smorlesi A, Acciarini S, Mondini E, Colleluori G, Pirazzini C, Kwiatkowska KM, Garagnani P, Franceschi C, Zingaretti MC, Dani C, Giordano A, Cinti S. Adipo-Epithelial Transdifferentiation in In Vitro Models of the Mammary Gland. Cells 2024; 13:943. [PMID: 38891075 PMCID: PMC11171678 DOI: 10.3390/cells13110943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Subcutaneous adipocytes are crucial for mammary gland epithelial development during pregnancy. Our and others' previous data have suggested that adipo-epithelial transdifferentiation could play a key role in the mammary gland alveolar development. In this study, we tested whether adipo-epithelial transdifferentiation occurs in vitro. Data show that, under appropriate co-culture conditions with mammary epithelial organoids (MEOs), mature adipocytes lose their phenotype and acquire an epithelial one. Interestingly, even in the absence of MEOs, extracellular matrix and diffusible growth factors are able to promote adipo-epithelial transdifferentiation. Gene and protein expression studies indicate that transdifferentiating adipocytes exhibit some characteristics of milk-secreting alveolar glands, including significantly higher expression of milk proteins such as whey acidic protein and β-casein. Similar data were also obtained in cultured human multipotent adipose-derived stem cell adipocytes. A miRNA sequencing experiment on the supernatant highlighted mir200c, which has a well-established role in the mesenchymal-epithelial transition, as a potential player in this phenomenon. Collectively, our data show that adipo-epithelial transdifferentiation can be reproduced in in vitro models where this phenomenon can be investigated at the molecular level.
Collapse
Affiliation(s)
- Jessica Perugini
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University—United Hospitals, 60126 Ancona, Italy; (J.P.); (A.S.); (S.A.); (E.M.); (G.C.); (M.C.Z.); (A.G.)
| | - Arianna Smorlesi
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University—United Hospitals, 60126 Ancona, Italy; (J.P.); (A.S.); (S.A.); (E.M.); (G.C.); (M.C.Z.); (A.G.)
| | - Samantha Acciarini
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University—United Hospitals, 60126 Ancona, Italy; (J.P.); (A.S.); (S.A.); (E.M.); (G.C.); (M.C.Z.); (A.G.)
| | - Eleonora Mondini
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University—United Hospitals, 60126 Ancona, Italy; (J.P.); (A.S.); (S.A.); (E.M.); (G.C.); (M.C.Z.); (A.G.)
| | - Georgia Colleluori
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University—United Hospitals, 60126 Ancona, Italy; (J.P.); (A.S.); (S.A.); (E.M.); (G.C.); (M.C.Z.); (A.G.)
| | - Chiara Pirazzini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy; (C.P.); (K.M.K.); (P.G.); (C.F.)
| | - Katarzyna Malgorzata Kwiatkowska
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy; (C.P.); (K.M.K.); (P.G.); (C.F.)
| | - Paolo Garagnani
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy; (C.P.); (K.M.K.); (P.G.); (C.F.)
- IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Claudio Franceschi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy; (C.P.); (K.M.K.); (P.G.); (C.F.)
- Laboratory of Systems Medicine of Healthy Aging, Institute of Biology and Biomedicine and Institute of Information Technology, Mathematics and Mechanics, Department of Applied Mathematics, N. I. Lobachevsky State University, 603005 Nizhny Novgorod, Russia
| | - Maria Cristina Zingaretti
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University—United Hospitals, 60126 Ancona, Italy; (J.P.); (A.S.); (S.A.); (E.M.); (G.C.); (M.C.Z.); (A.G.)
| | - Christian Dani
- Faculté de Médecine, CNRS, INSERM, iBV, Université Côte d’Azur, CEDEX 2, F-06107 Nice, France;
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University—United Hospitals, 60126 Ancona, Italy; (J.P.); (A.S.); (S.A.); (E.M.); (G.C.); (M.C.Z.); (A.G.)
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University—United Hospitals, 60126 Ancona, Italy; (J.P.); (A.S.); (S.A.); (E.M.); (G.C.); (M.C.Z.); (A.G.)
| |
Collapse
|
20
|
Nomura K, Kimira Y, Kobayashi R, Shiobara Y, Osawa Y, Kataoka-Matsushita A, Shimizu J, Wada M, Mano H. Collagen-derived dipeptide prolyl-hydroxyproline cooperates with Foxg1 to activate the PGC-1α promoter and induce brown adipocyte-like phenotype in rosiglitazone-treated C3H10T1/2 cells. Front Nutr 2024; 11:1375532. [PMID: 38812940 PMCID: PMC11133597 DOI: 10.3389/fnut.2024.1375532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/23/2024] [Indexed: 05/31/2024] Open
Abstract
Background The global obesity epidemic is a significant public health issue, often leading to metabolic disorders such as diabetes and cardiovascular diseases. Collagen peptides (CP) and their bioactive component, Prolyl-hydroxyproline (Pro-Hyp), have shown potential in reducing adipocyte size, with unclear mechanisms concerning brown adipocyte differentiation. Methods We investigated the effects of Pro-Hyp on the differentiation of brown adipocytes in C3H10T1/2 mesenchymal stem cells, focusing on its impact on adipocyte size, gene expression related to brown fat function, and mitochondrial activity. Results Pro-Hyp treatment decreased adipocyte size and upregulated brown fat-specific genes, including C/EBPα, PGC-1α, and UCP-1. Remarkably, it did not alter PPARγ expression. Pro-Hyp also elevated mitochondrial activity, suggesting enhanced brown adipocyte functionality. A Pro-Hyp responsive element was identified in the PGC-1α gene promoter, which facilitated the binding of the Foxg1 transcription factor, indicating a novel regulatory mechanism. Conclusion Pro-Hyp promotes brown adipocyte differentiation, potentially offering a therapeutic strategy for obesity management. This study provides a molecular basis for the anti-obesity effects of CP, although further in vivo studies are needed to confirm these findings and to investigate the potential impact on beige adipocyte differentiation.
Collapse
Affiliation(s)
- Kaho Nomura
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
- Department of Molecular Microbiology, Faculty of Life Science, Tokyo University of Agriculture, Tokyo, Japan
| | - Yoshifumi Kimira
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Ryosuke Kobayashi
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Yuna Shiobara
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Yoshihiro Osawa
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| | | | - Jun Shimizu
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Masahiro Wada
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Hiroshi Mano
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| |
Collapse
|
21
|
Iacobini C, Vitale M, Haxhi J, Menini S, Pugliese G. Impaired Remodeling of White Adipose Tissue in Obesity and Aging: From Defective Adipogenesis to Adipose Organ Dysfunction. Cells 2024; 13:763. [PMID: 38727299 PMCID: PMC11083890 DOI: 10.3390/cells13090763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
The adipose organ adapts and responds to internal and environmental stimuli by remodeling both its cellular and extracellular components. Under conditions of energy surplus, the subcutaneous white adipose tissue (WAT) is capable of expanding through the enlargement of existing adipocytes (hypertrophy), followed by de novo adipogenesis (hyperplasia), which is impaired in hypertrophic obesity. However, an impaired hyperplastic response may result from various defects in adipogenesis, leading to different WAT features and metabolic consequences, as discussed here by reviewing the results of the studies in animal models with either overexpression or knockdown of the main molecular regulators of the two steps of the adipogenesis process. Moreover, impaired WAT remodeling with aging has been associated with various age-related conditions and reduced lifespan expectancy. Here, we delve into the latest advancements in comprehending the molecular and cellular processes underlying age-related changes in WAT function, their involvement in common aging pathologies, and their potential as therapeutic targets to influence both the health of elderly people and longevity. Overall, this review aims to encourage research on the mechanisms of WAT maladaptation common to conditions of both excessive and insufficient fat tissue. The goal is to devise adipocyte-targeted therapies that are effective against both obesity- and age-related disorders.
Collapse
|
22
|
Bellitto V, Gabrielli MG, Martinelli I, Roy P, Nittari G, Cocci P, Palermo FA, Amenta F, Micioni Di Bonaventura MV, Cifani C, Tomassoni D, Tayebati SK. Dysfunction of the Brown Adipose Organ in HFD-Obese Rats and Effect of Tart Cherry Supplementation. Antioxidants (Basel) 2024; 13:388. [PMID: 38671836 PMCID: PMC11047636 DOI: 10.3390/antiox13040388] [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: 02/09/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Obesity has a great impact on adipose tissue biology, based on its function as a master regulator of energy balance. Brown adipose tissue (BAT) undergoes remodeling, and its activity declines in obese subjects due to a whitening process. The anti-obesity properties of fruit extracts have been reported. The effects of tart cherry against oxidative stress, inflammation, and the whitening process in the BAT of obese rats were investigated. Intrascapular BAT (iBAT) alterations and effects of Prunus cerasus L. were debated in rats fed for 17 weeks with a high-fat diet (DIO), in DIO supplemented with seed powder (DS), and with seed powder plus the juice (DJS) of tart cherry compared to CHOW rats fed with a normo-caloric diet. iBAT histologic observations revealed a whitening process in DIO rats that was reduced in the DS and DJS groups. A modulation of uncoupling protein-1 (UCP-1) protein and gene expression specifically were detected in the obese phenotype. An upregulation of UCP-1 and related thermogenic genes after tart cherry intake was detected compared to the DIO group. Metabolic adjustment, endoplasmic reticulum stress, protein carbonylation, and the inflammatory microenvironment in the iBAT were reported in DIO rats. The analysis demonstrated an iBAT modulation that tart cherry promoted. In addition to our previous results, these data confirm the protective impact of tart cherry consumption on obesity.
Collapse
Affiliation(s)
- Vincenzo Bellitto
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Maria Gabriella Gabrielli
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (M.G.G.); (P.C.); (F.A.P.); (D.T.)
| | - Ilenia Martinelli
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Proshanta Roy
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Giulio Nittari
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Paolo Cocci
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (M.G.G.); (P.C.); (F.A.P.); (D.T.)
| | - Francesco Alessandro Palermo
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (M.G.G.); (P.C.); (F.A.P.); (D.T.)
| | - Francesco Amenta
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Maria Vittoria Micioni Di Bonaventura
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Carlo Cifani
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (M.G.G.); (P.C.); (F.A.P.); (D.T.)
| | - Seyed Khosrow Tayebati
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| |
Collapse
|
23
|
Jung KM, Lin L, Piomelli D. Overactivation of the Endocannabinoid System in Adolescence Disrupts Adult Adipose Organ Function in Mice. Cells 2024; 13:461. [PMID: 38474425 PMCID: PMC10930932 DOI: 10.3390/cells13050461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024] Open
Abstract
Cannabis use stimulates calorie intake, but epidemiological studies show that people who regularly use it are leaner than those who don't. Two explanations have been proposed for this paradoxical finding. One posits that Δ9-tetrahydrocannabinol (THC) in cannabis desensitizes adipose CB1 cannabinoid receptors, stopping their stimulating effects on lipogenesis and adipogenesis. Another explanation is that THC exposure in adolescence, when habitual cannabis use typically starts, produces lasting changes in the developing adipose organ, which impacts adult systemic energy use. Here, we consider these possibilities in the light of a study which showed that daily THC administration in adolescent mice produces an adult metabolic phenotype characterized by reduced fat mass, partial resistance to obesity and dyslipidemia, and impaired thermogenesis and lipolysis. The phenotype, whose development requires activation of CB1 receptors in differentiated adipocytes, is associated with overexpression of myocyte proteins in the adipose organ with unchanged CB1 expression. We propose that adolescent exposure to THC causes lasting adipocyte dysfunction and the consequent emergence of a metabolic state that only superficially resembles healthy leanness. A corollary of this hypothesis, which should be addressed in future studies, is that CB1 receptors and their endocannabinoid ligands may contribute to the maintenance of adipocyte differentiation during adolescence.
Collapse
Affiliation(s)
- Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697, USA; (K.-M.J.); (L.L.)
| | - Lin Lin
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697, USA; (K.-M.J.); (L.L.)
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697, USA; (K.-M.J.); (L.L.)
- Department of Biological Chemistry, University of California, Irvine, CA 92697, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA
| |
Collapse
|
24
|
Pan T, Lee YM, Takimoto E, Ueda K, Liu PY, Shen HH. Inhibitory effects of naringenin on estrogen deficiency-induced obesity via regulation of mitochondrial dynamics and AMPK activation associated with white adipose tissue browning. Life Sci 2024; 340:122453. [PMID: 38272439 DOI: 10.1016/j.lfs.2024.122453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024]
Abstract
AIMS Post-ovariectomy (OVX) changes in hormones induce obesity and white adipose tissue (WAT) inflammation. Increased energy expenditure via WAT browning is a novel therapeutic strategy for treating obesity. Naringenin (NAR) reduces inflammation and lipogenesis in obesity and attenuates estrogen deficiency-associated metabolic disorders; however, its role in WAT browning remains unclear. MATERIALS AND METHODS We investigated NAR ability to inhibit estrogen deficiency-associated obesity in vivo using a rat model and in vitro using 3T3-L1 adipocytes. KEY FINDINGS NAR significantly decreased the body weight and WAT mass of rats. O2 consumption, CO2 production, and energy expenditure were significantly lower in the OVX group than in the sham group, but NAR treatment reversed these effects of OVX. NAR treatment markedly improved glucose intolerance and lipid profiles as well as leptin, adiponectin, and irisin levels. NAR upregulated markers of browning and mitochondrial biogenesis in inguinal WAT. Moreover, it enhanced markers of mitochondrial fusion and inhibited fission via activating the AMP-activated protein kinase pathway. Similar results were observed in 3T3-L1 adipocytes. Moreover, NAR-induced mitochondrial biogenesis and fusion were suppressed by dorsomorphin (an AMP-activated protein kinase inhibitor). SIGNIFICANCE NAR alleviates obesity and metabolic dysfunction through the induction of WAT browning achieved via the modulation of AMP-activated protein kinase-regulated mitochondrial dynamics in WATs. NAR supplementation may therefore represent a potential intervention for preventing postmenopausal adipose tissue dysregulation.
Collapse
Affiliation(s)
- Tong Pan
- Graduate Institute and Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Yen-Mei Lee
- Graduate Institute and Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kazutaka Ueda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Pang-Yen Liu
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Hsin-Hsueh Shen
- Graduate Institute and Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
25
|
Ren W, Hua M, Cao F, Zeng W. The Sympathetic-Immune Milieu in Metabolic Health and Diseases: Insights from Pancreas, Liver, Intestine, and Adipose Tissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306128. [PMID: 38039489 PMCID: PMC10885671 DOI: 10.1002/advs.202306128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/28/2023] [Indexed: 12/03/2023]
Abstract
Sympathetic innervation plays a crucial role in maintaining energy balance and contributes to metabolic pathophysiology. Recent evidence has begun to uncover the innervation landscape of sympathetic projections and sheds light on their important functions in metabolic activities. Additionally, the immune system has long been studied for its essential roles in metabolic health and diseases. In this review, the aim is to provide an overview of the current research progress on the sympathetic regulation of key metabolic organs, including the pancreas, liver, intestine, and adipose tissues. In particular, efforts are made to highlight the critical roles of the peripheral nervous system and its potential interplay with immune components. Overall, it is hoped to underscore the importance of studying metabolic organs from a comprehensive and interconnected perspective, which will provide valuable insights into the complex mechanisms underlying metabolic regulation and may lead to novel therapeutic strategies for metabolic diseases.
Collapse
Affiliation(s)
- Wenran Ren
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
| | - Meng Hua
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
| | - Fang Cao
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhou563000China
| | - Wenwen Zeng
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
- SXMU‐Tsinghua Collaborative Innovation Center for Frontier MedicineTaiyuan030001China
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijing100084China
| |
Collapse
|
26
|
Behrooz AB, Cordani M, Fiore A, Donadelli M, Gordon JW, Klionsky DJ, Ghavami S. The obesity-autophagy-cancer axis: Mechanistic insights and therapeutic perspectives. Semin Cancer Biol 2024; 99:24-44. [PMID: 38309540 DOI: 10.1016/j.semcancer.2024.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/19/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Autophagy, a self-degradative process vital for cellular homeostasis, plays a significant role in adipose tissue metabolism and tumorigenesis. This review aims to elucidate the complex interplay between autophagy, obesity, and cancer development, with a specific emphasis on how obesity-driven changes affect the regulation of autophagy and subsequent implications for cancer risk. The burgeoning epidemic of obesity underscores the relevance of this research, particularly given the established links between obesity, autophagy, and various cancers. Our exploration delves into hormonal influence, notably INS (insulin) and LEP (leptin), on obesity and autophagy interactions. Further, we draw attention to the latest findings on molecular factors linking obesity to cancer, including hormonal changes, altered metabolism, and secretory autophagy. We posit that targeting autophagy modulation may offer a potent therapeutic approach for obesity-associated cancer, pointing to promising advancements in nanocarrier-based targeted therapies for autophagy modulation. However, we also recognize the challenges inherent to these approaches, particularly concerning their precision, control, and the dual roles autophagy can play in cancer. Future research directions include identifying novel biomarkers, refining targeted therapies, and harmonizing these approaches with precision medicine principles, thereby contributing to a more personalized, effective treatment paradigm for obesity-mediated cancer.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, University of Manitoba, College of Medicine, Winnipeg, Manitoba, Canada; Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Alessandra Fiore
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Joseph W Gordon
- Department of Human Anatomy and Cell Science, University of Manitoba, College of Medicine, Winnipeg, Manitoba, Canada; Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Saeid Ghavami
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA; Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada; Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
27
|
Li X, Ren H, Xu H, Han X, Lu J, Yang Z. Behind BMI: The Potential Indicative Role of Abdominal Ectopic Fat on Glucose Metabolism. Obes Facts 2024; 17:158-168. [PMID: 38246158 PMCID: PMC10987183 DOI: 10.1159/000536160] [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: 10/11/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
INTRODUCTION The purpose of this study was to compare the difference in abdominal fat distribution between different metabolic groups and find the ectopic fat with the most risk significance. METHODS A total of 98 subjects were enrolled; there were 53 cases in the normal glucose metabolism group and 45 cases in the abnormal glucose metabolism group. Chemical shift-encoded magnetic resonance imaging was applied for quantification of pancreatic fat fraction (PFF) and hepatic fat fraction (HFF), subcutaneous adipose tissue (SAT), and visceral adipose tissue (VAT). The correlation and the difference of fat distribution between different metabolism groups were analyzed. The receiver operating characteristic (ROC) curve was used to analyze the suggestive effect of different body fat fraction. RESULTS Correlation analysis showed that body mass index (BMI) had the strongest correlation with fasting insulin (r = 0.473, p < 0.001), HOMA-IR (r = 0.363, p < 0.001), and C-reactive protein (r = 0.245, p < 0.05). Pancreatic fat has a good correlation with fasting blood glucose (r = 0.247, p < 0.05) and HbA1c (r = 0.363, p < 0.001). With the increase of BMI, PFF, VAT, and SAT showed a clear upward trend, but liver fat was distributed relatively more randomly. The pancreatic fat content in the abnormal glucose metabolism group is significantly higher than that in the normal group, and pancreatic fat is also a reliable indicator of abnormal glucose metabolism, especially in the normal and overweight groups (the area under the curve was 0.859 and 0.864, respectively). CONCLUSION MR-based fat quantification techniques can provide additional information on fat distribution. There are differences in fat distribution among people with different metabolic status. People with more severe pancreatic fat deposition have a higher risk of glucose metabolism disorders.
Collapse
Affiliation(s)
- Xiaoyang Li
- Beijing Friendship Hospital, Capital Medical University, Beijing, China,
| | - Hao Ren
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hui Xu
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xinjun Han
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jun Lu
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhenghan Yang
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Da Rosa SE, Neves EB, Martinez EC, Marson RA, Reis VM. Subcutaneous and Visceral Fat: Relation with Brown Adipose Tissue Activation in Women. Sports Med Int Open 2024; 8:a21876974. [PMID: 38312927 PMCID: PMC10832575 DOI: 10.1055/a-2187-6974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/25/2023] [Indexed: 02/06/2024] Open
Abstract
Brown adipose tissue (BAT) helps control body weight and is inversely correlated with body fat, but it is unclear whether it is subcutaneous adipose tissue (SAT) or visceral adipose tissue (VAT) that is related to BAT activation. The presented study aimed to verify the relation of SAT and VAT on BAT activation through infrared thermography (IRT) and cold stimulation in adult women. Forty women were evaluated in body composition and skin temperature (Tskin) acquisition by IRT. Student's independent t-test, Pearson's correlation, and two-way repeated measures ANOVA with Tukey post-hoc were applied. Women with low amounts of SAT and VAT had a significant increase in supraclavicular Tskin (SCVT). Medium negative degrees of linear variation were found before and after cold stimulation between SCVT, SAT and VAT. A significant effect of the moment factor and the group factor on the SCVT between subjects divided into the groups were pointed out. No difference was found in the relation between SAT, VAT, and BAT in adult women, pointing out that both types of fat are equally related. These results can help clinical practice understand clearly, through IRT, that the high accumulation of SAT and VAT can impair the activation of BAT and hinder the loss of weight in women.
Collapse
Affiliation(s)
- Samir Ezequiel Da Rosa
- Research Center in Sports Sciences, Health Sciences and Human
Development (CIDESD), Trás-os-Montes e Alto Douro University, UTAD, Vila
Real, Portugal
| | - Eduardo Borba Neves
- Programa de Pós-Graduação em engenharia
Biomédica, Universidade Tecnológica Federal do Paraná,
Curitiba, Brazil
| | - Eduardo Camilo Martinez
- Research Center in Sports Sciences, Health Sciences and Human
Development (CIDESD), Trás-os-Montes e Alto Douro University, UTAD, Vila
Real, Portugal
| | - Runer Augusto Marson
- Seção de Apoio à Operacionalidade, Brazilian
Army Physical Training Research Institute (IPCFEx), Rio de Janeiro,
Brazil
| | - Victor Machado Reis
- Research Center in Sports Sciences, Health Sciences and Human
Development (CIDESD), Trás-os-Montes e Alto Douro University, UTAD, Vila
Real, Portugal
| |
Collapse
|
29
|
Pfeifer A, Mikhael M, Niemann B. Inosine: novel activator of brown adipose tissue and energy homeostasis. Trends Cell Biol 2024; 34:72-82. [PMID: 37188562 DOI: 10.1016/j.tcb.2023.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/17/2023]
Abstract
Extracellular purinergic molecules act as signaling molecules that bind to cellular receptors and regulate signaling pathways. Growing evidence suggests that purines regulate adipocyte function and whole-body metabolism. Here, we focus on one specific purine: inosine. Brown adipocytes, which are important regulators of whole-body energy expenditure (EE), release inosine when they are stressed or become apoptotic. Unexpectedly, inosine activates EE in neighboring brown adipocytes and enhances differentiation of brown preadipocytes. Increasing extracellular inosine, either directly by increasing inosine intake or indirectly via pharmacological inhibition of cellular inosine transporters, increases whole-body EE and counteracts obesity. Thus, inosine and other closely related purines might be a novel approach to tackle obesity and associated metabolic disorders by enhancing EE.
Collapse
Affiliation(s)
- Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany.
| | - Mickel Mikhael
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Birte Niemann
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| |
Collapse
|
30
|
Pelczyńska M, Miller-Kasprzak E, Piątkowski M, Mazurek R, Klause M, Suchecka A, Bucoń M, Bogdański P. The Role of Adipokines and Myokines in the Pathogenesis of Different Obesity Phenotypes-New Perspectives. Antioxidants (Basel) 2023; 12:2046. [PMID: 38136166 PMCID: PMC10740719 DOI: 10.3390/antiox12122046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/19/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
Obesity is a characteristic disease of the twenty-first century that is affecting an increasing percentage of society. Obesity expresses itself in different phenotypes: normal-weight obesity (NWO), metabolically obese normal-weight (MONW), metabolically healthy obesity (MHO), and metabolically unhealthy obesity (MUO). A range of pathophysiological mechanisms underlie the occurrence of obesity, including inflammation, oxidative stress, adipokine secretion, and other processes related to the pathophysiology of adipose tissue (AT). Body mass index (BMI) is the key indicator in the diagnosis of obesity; however, in the case of the NWO and MONW phenotypes, the metabolic disturbances are present despite BMI being within the normal range. On the other hand, MHO subjects with elevated BMI values do not present metabolic abnormalities. The MUO phenotype involves both a high BMI value and an abnormal metabolic profile. In this regard, attention has been focused on the variety of molecules produced by AT and their role in the development of obesity. Nesfatin-1, neuregulin 4, myonectin, irisin, and brain-derived neurotrophic factor (BDNF) all seem to have protective effects against obesity. The primary mechanism underlying the action of nesfatin-1 involves an increase in insulin sensitivity and reduced food intake. Neuregulin 4 sup-presses lipogenesis, decreases lipid accumulation, and reduces chronic low-grade inflammation. Myonectin lowers the amount of fatty acids in the bloodstream by increasing their absorption in the liver and AT. Irisin stimulates the browning of white adipose tissue (WAT) and consequently in-creases energy expenditure, additionally regulating glucose metabolism. Another molecule, BDNF, has anorexigenic effects. Decorin protects against the development of hyperglycemia, but may also contribute to proinflammatory processes. Similar effects are shown in the case of visfatin and chemerin, which may predispose to obesity. Visfatin increases adipogenesis, causes cholesterol accumulation in macrophages, and contributes to the development of glucose intolerance. Chemerin induces angiogenesis, which promotes the expansion of AT. This review aims to discuss the role of adipokines and myokines in the pathogenesis of the different obesity phenotypes.
Collapse
Affiliation(s)
- Marta Pelczyńska
- Chair and Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, 84 Szamarzewskiego Street, 60-569 Poznań, Poland; (E.M.-K.); (P.B.)
| | - Ewa Miller-Kasprzak
- Chair and Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, 84 Szamarzewskiego Street, 60-569 Poznań, Poland; (E.M.-K.); (P.B.)
| | - Marcin Piątkowski
- Faculty of Medicine, Poznan University of Medical Sciences, 70 Bukowska Street, 60-812 Poznań, Poland
| | - Roksana Mazurek
- Faculty of Medicine, Poznan University of Medical Sciences, 70 Bukowska Street, 60-812 Poznań, Poland
| | - Mateusz Klause
- Faculty of Medicine, Poznan University of Medical Sciences, 70 Bukowska Street, 60-812 Poznań, Poland
| | - Anna Suchecka
- Faculty of Medicine, Poznan University of Medical Sciences, 70 Bukowska Street, 60-812 Poznań, Poland
| | - Magdalena Bucoń
- Faculty of Medicine, Poznan University of Medical Sciences, 70 Bukowska Street, 60-812 Poznań, Poland
| | - Paweł Bogdański
- Chair and Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, 84 Szamarzewskiego Street, 60-569 Poznań, Poland; (E.M.-K.); (P.B.)
| |
Collapse
|
31
|
Gyurina K, Yarmak M, Sasi-Szabó L, Molnár S, Méhes G, Röszer T. Loss of Uncoupling Protein 1 Expression in the Subcutaneous Adipose Tissue Predicts Childhood Obesity. Int J Mol Sci 2023; 24:16706. [PMID: 38069028 PMCID: PMC10706300 DOI: 10.3390/ijms242316706] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Stimulation of thermogenesis by inducing uncoupling protein 1 (UCP1) expression in adipocytes is thought to promote weight loss by increasing energy expenditure, and it is postulated that the human newborn has thermogenic subcutaneous fat depots. However, it remains unclear whether a relevant number of UCP1-expressing (UCP1+) adipocytes exist in the early postnatal life. Here we studied the distribution of UCP1 and the expression of thermogenic genes in the subcutaneous adipose tissues of the human fetus, infant and child. We show that the deep layer of human fetal and neonatal subcutaneous fat, particularly the abdominal wall, is rich in UCP1+ adipocytes. These adipocytes develop in the late third trimester and persist throughout childhood, expressing a panel of genes linked to mitochondrial biogenesis and thermogenesis. During the early childhood adiposity rebound-a critical phase that determines obesity risk later in life-the absence of adipose tissue UCP1 expression in children with normal body mass index (BMI) correlates with an obesity-associated gene expression signature. Finally, UCP1 expression is negatively correlated with BMI z-score and adipocyte size in infants and children. Overall, our results show that the absence of UCP1 expression in adipose tissue is an early indicator of adipose tissue expansion in children.
Collapse
Affiliation(s)
- Katalin Gyurina
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
| | - Mariia Yarmak
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
| | - László Sasi-Szabó
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
| | - Sarolta Molnár
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (S.M.)
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (S.M.)
| | - Tamás Röszer
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
- Institute of Neurobiology, Ulm University, 89081 Ulm, Germany
| |
Collapse
|
32
|
Lu S, Cao ZB. Interplay between Vitamin D and Adipose Tissue: Implications for Adipogenesis and Adipose Tissue Function. Nutrients 2023; 15:4832. [PMID: 38004226 PMCID: PMC10675652 DOI: 10.3390/nu15224832] [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: 10/19/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Adipose tissue encompasses various types, including White Adipose Tissue (WAT), Brown Adipose Tissue (BAT), and beige adipose tissue, each having distinct roles in energy storage and thermogenesis. Vitamin D (VD), a fat-soluble vitamin, maintains a complex interplay with adipose tissue, exerting significant effects through its receptor (VDR) on the normal development and functioning of adipocytes. The VDR and associated metabolic enzymes are widely expressed in the adipocytes of both rodents and humans, and they partake in the regulation of fat metabolism and functionality through various pathways. These encompass adipocyte differentiation, adipogenesis, inflammatory responses, and adipokine synthesis and secretion. This review primarily appraises the role and mechanisms of VD in different adipocyte differentiation, lipid formation, and inflammatory responses, concentrating on the pivotal role of the VD/VDR pathway in adipogenesis. This insight furnishes new perspectives for the development of micronutrient-related intervention strategies in the prevention and treatment of obesity.
Collapse
Affiliation(s)
| | - Zhen-Bo Cao
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China;
| |
Collapse
|
33
|
Schena E, Mattioli E, Peres C, Zanotti L, Morselli P, Iozzo P, Guzzardi MA, Bernardini C, Forni M, Nesci S, Caprio M, Cecchetti C, Pagotto U, Gabusi E, Cattini L, Lisignoli G, Blalock W, Gambineri A, Lattanzi G. Mineralocorticoid Receptor Antagonism Prevents Type 2 Familial Partial Lipodystrophy Brown Adipocyte Dysfunction. Cells 2023; 12:2586. [PMID: 37998321 PMCID: PMC10670260 DOI: 10.3390/cells12222586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/29/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023] Open
Abstract
Type-2 Familial Partial Lipodystrophy (FPLD2), a rare lipodystrophy caused by LMNA mutations, is characterized by a loss of subcutaneous fat from the trunk and limbs and excess accumulation of adipose tissue in the neck and face. Several studies have reported that the mineralocorticoid receptor (MR) plays an essential role in adipose tissue differentiation and functionality. We previously showed that brown preadipocytes isolated from a FPLD2 patient's neck aberrantly differentiate towards the white lineage. As this condition may be related to MR activation, we suspected altered MR dynamics in FPLD2. Despite cytoplasmic MR localization in control brown adipocytes, retention of MR was observed in FPLD2 brown adipocyte nuclei. Moreover, overexpression of wild-type or mutated prelamin A caused GFP-MR recruitment to the nuclear envelope in HEK293 cells, while drug-induced prelamin A co-localized with endogenous MR in human preadipocytes. Based on in silico analysis and in situ protein ligation assays, we could suggest an interaction between prelamin A and MR, which appears to be inhibited by mineralocorticoid receptor antagonism. Importantly, the MR antagonist spironolactone redirected FPLD2 preadipocyte differentiation towards the brown lineage, avoiding the formation of enlarged and dysmorphic lipid droplets. Finally, beneficial effects on brown adipose tissue activity were observed in an FPLD2 patient undergoing spironolactone treatment. These findings identify MR as a new lamin A interactor and a new player in lamin A-linked lipodystrophies.
Collapse
Affiliation(s)
- Elisa Schena
- Unit of Bologna, CNR—National Research Council of Italy, Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, 40136 Bologna, Italy; (E.S.); (E.M.); (C.P.); (W.B.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Elisabetta Mattioli
- Unit of Bologna, CNR—National Research Council of Italy, Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, 40136 Bologna, Italy; (E.S.); (E.M.); (C.P.); (W.B.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Chiara Peres
- Unit of Bologna, CNR—National Research Council of Italy, Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, 40136 Bologna, Italy; (E.S.); (E.M.); (C.P.); (W.B.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Laura Zanotti
- Unit of Gynecology and Obstetrics, Division of Endocrinology and Diabetes Prevention and Care, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.Z.); (C.C.); (U.P.); (A.G.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Paolo Morselli
- Plastic Surgery Unit, Department of Specialised, Experimental and Diagnostic Medicine, Alma Mater Studiorum University of Bologna, S. Orsola-Malpighi Hospital, 40126 Bologna, Italy;
| | - Patricia Iozzo
- CNR—National Research Council of Italy, Institute of Clinical Physiology, 56124 Pisa, Italy; (P.I.); (M.A.G.)
| | - Maria Angela Guzzardi
- CNR—National Research Council of Italy, Institute of Clinical Physiology, 56124 Pisa, Italy; (P.I.); (M.A.G.)
| | - Chiara Bernardini
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Ozzano Emilia, Italy; (C.B.); (S.N.)
| | - Monica Forni
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Salvatore Nesci
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Ozzano Emilia, Italy; (C.B.); (S.N.)
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele, 00163 Rome, Italy;
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Carolina Cecchetti
- Unit of Gynecology and Obstetrics, Division of Endocrinology and Diabetes Prevention and Care, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.Z.); (C.C.); (U.P.); (A.G.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Uberto Pagotto
- Unit of Gynecology and Obstetrics, Division of Endocrinology and Diabetes Prevention and Care, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.Z.); (C.C.); (U.P.); (A.G.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Elena Gabusi
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (E.G.); (L.C.); (G.L.)
| | - Luca Cattini
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (E.G.); (L.C.); (G.L.)
| | - Gina Lisignoli
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (E.G.); (L.C.); (G.L.)
| | - William Blalock
- Unit of Bologna, CNR—National Research Council of Italy, Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, 40136 Bologna, Italy; (E.S.); (E.M.); (C.P.); (W.B.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Alessandra Gambineri
- Unit of Gynecology and Obstetrics, Division of Endocrinology and Diabetes Prevention and Care, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.Z.); (C.C.); (U.P.); (A.G.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Giovanna Lattanzi
- Unit of Bologna, CNR—National Research Council of Italy, Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, 40136 Bologna, Italy; (E.S.); (E.M.); (C.P.); (W.B.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| |
Collapse
|
34
|
Panico G, Fasciolo G, Migliaccio V, De Matteis R, Lionetti L, Napolitano G, Agnisola C, Venditti P, Lombardi A. 1,3-Butanediol Administration Increases β-Hydroxybutyrate Plasma Levels and Affects Redox Homeostasis, Endoplasmic Reticulum Stress, and Adipokine Production in Rat Gonadal Adipose Tissue. Antioxidants (Basel) 2023; 12:1471. [PMID: 37508009 PMCID: PMC10376816 DOI: 10.3390/antiox12071471] [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: 06/19/2023] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Ketone bodies (KBs) are an alternative energy source under starvation and play multiple roles as signaling molecules regulating energy and metabolic homeostasis. The mechanism by which KBs influence visceral white adipose tissue physiology is only partially known, and our study aimed to shed light on the effects they exert on such tissue. To this aim, we administered 1,3-butanediol (BD) to rats since it rapidly enhances β-hydroxybutyrate serum levels, and we evaluated the effect it induces within 3 h or after 14 days of treatment. After 14 days of treatment, rats showed a decrease in body weight gain, energy intake, gonadal-WAT (gWAT) weight, and adipocyte size compared to the control. BD exerted a pronounced antioxidant effect and directed redox homeostasis toward reductive stress, already evident within 3 h after its administration. BD lowered tissue ROS levels and oxidative damage to lipids and proteins and enhanced tissue soluble and enzymatic antioxidant capacity as well as nuclear erythroid factor-2 protein levels. BD also reduced specific mitochondrial maximal oxidative capacity and induced endoplasmic reticulum stress as well as interrelated processes, leading to changes in the level of adipokines/cytokines involved in inflammation, macrophage infiltration into gWAT, adipocyte differentiation, and lipolysis.
Collapse
Affiliation(s)
- Giuliana Panico
- Department of Biology, University of Naples Federico II, Complesso Monte Sant'Angelo Via Cintia 26, 80126 Napoli, Italy
| | - Gianluca Fasciolo
- Department of Biology, University of Naples Federico II, Complesso Monte Sant'Angelo Via Cintia 26, 80126 Napoli, Italy
| | - Vincenzo Migliaccio
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Rita De Matteis
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Lillà Lionetti
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Gaetana Napolitano
- Department of Science and Technology, Parthenope University of Naples, 80143 Naples, Italy
| | - Claudio Agnisola
- Department of Biology, University of Naples Federico II, Complesso Monte Sant'Angelo Via Cintia 26, 80126 Napoli, Italy
| | - Paola Venditti
- Department of Biology, University of Naples Federico II, Complesso Monte Sant'Angelo Via Cintia 26, 80126 Napoli, Italy
| | - Assunta Lombardi
- Department of Biology, University of Naples Federico II, Complesso Monte Sant'Angelo Via Cintia 26, 80126 Napoli, Italy
| |
Collapse
|
35
|
Cinti S. Obese Adipocytes Have Altered Redox Homeostasis with Metabolic Consequences. Antioxidants (Basel) 2023; 12:1449. [PMID: 37507987 PMCID: PMC10376822 DOI: 10.3390/antiox12071449] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
White and brown adipose tissues are organized to form a real organ, the adipose organ, in mice and humans. White adipocytes of obese animals and humans are hypertrophic. This condition is accompanied by a series of organelle alterations and stress of the endoplasmic reticulum. This stress is mainly due to reactive oxygen species activity and accumulation, lending to NLRP3 inflammasome activation. This last causes death of adipocytes by pyroptosis and the formation of large cellular debris that must be removed by macrophages. During their chronic scavenging activity, macrophages produce several secretory products that have collateral consequences, including interference with insulin receptor activity, causing insulin resistance. The latter is accompanied by an increased noradrenergic inhibitory innervation of Langerhans islets with de-differentiation of beta cells and type 2 diabetes. The whitening of brown adipocytes could explain the different critical death size of visceral adipocytes and offer an explanation for the worse clinical consequence of visceral fat accumulation. White to brown transdifferentiation has been proven in mice and humans. Considering the energy-dispersing activity of brown adipose tissue, transdifferentiation opens new therapeutic perspectives for obesity and related disorders.
Collapse
Affiliation(s)
- Saverio Cinti
- Scientific Director Centre of Obesity, Marche Polytechnic University, Via Tronto 10a, 60126 Ancona, Italy
| |
Collapse
|
36
|
Lin L, Jung KM, Lee HL, Le J, Colleluori G, Wood C, Palese F, Squire E, Ramirez J, Su S, Torrens A, Fotio Y, Tang L, Yu C, Yang Q, Huang L, DiPatrizio N, Jang C, Cinti S, Piomelli D. Adolescent exposure to low-dose THC disrupts energy balance and adipose organ homeostasis in adulthood. Cell Metab 2023; 35:1227-1241.e7. [PMID: 37267956 PMCID: PMC10524841 DOI: 10.1016/j.cmet.2023.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 02/07/2023] [Accepted: 05/05/2023] [Indexed: 06/04/2023]
Abstract
One of cannabis' most iconic effects is the stimulation of hedonic high-calorie eating-the "munchies"-yet habitual cannabis users are, on average, leaner than non-users. We asked whether this phenotype might result from lasting changes in energy balance established during adolescence, when use of the drug often begins. We found that daily low-dose administration of cannabis' intoxicating constituent, Δ9-tetrahydrocannabinol (THC), to adolescent male mice causes an adult metabolic phenotype characterized by reduced fat mass, increased lean mass and utilization of fat as fuel, partial resistance to diet-induced obesity and dyslipidemia, enhanced thermogenesis, and impaired cold- and β-adrenergic receptor-stimulated lipolysis. Further analyses revealed that this phenotype is associated with molecular anomalies in the adipose organ, including ectopic overexpression of muscle-associated proteins and heightened anabolic processing. Thus, adolescent exposure to THC may promote an enduring "pseudo-lean" state that superficially resembles healthy leanness but might in fact be rooted in adipose organ dysfunction.
Collapse
Affiliation(s)
- Lin Lin
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Hye-Lim Lee
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Johnny Le
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Georgia Colleluori
- Department of Clinical and Experimental Medicine, Center of Obesity, Marche Polytechnic University, Ancona 600126, Italy
| | - Courtney Wood
- Department of Biomedical Sciences, University of California, Riverside, Riverside, CA 92697, USA
| | - Francesca Palese
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Erica Squire
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Jade Ramirez
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Shiqi Su
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Alexa Torrens
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Yannick Fotio
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Lingyi Tang
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Clinton Yu
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Qin Yang
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Lan Huang
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Nicholas DiPatrizio
- Department of Biomedical Sciences, University of California, Riverside, Riverside, CA 92697, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Saverio Cinti
- Department of Clinical and Experimental Medicine, Center of Obesity, Marche Polytechnic University, Ancona 600126, Italy
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
37
|
Kesić M, Baković P, Farkaš V, Bagarić R, Kolarić D, Štefulj J, Čičin-Šain L. Constitutive Serotonin Tone as a Modulator of Brown Adipose Tissue Thermogenesis: A Rat Study. Life (Basel) 2023; 13:1436. [PMID: 37511811 PMCID: PMC10381595 DOI: 10.3390/life13071436] [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: 05/04/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Brown adipose tissue (BAT), an important regulator of thermogenic and metabolic processes, is considered a promising target to combat metabolic disorders. The neurotransmitter and hormone serotonin (5HT) is a major modulator of energy homeostasis, with its central and peripheral pools acting in opposing ways. To better understand how individual variations in 5HT homeostasis influence the thermogenic functionality of BAT, we used a rat model consisting of two sublines with constitutively increased (high-5HT) or decreased (low-5HT) whole-body 5HT tone, developed by selective breeding for platelet 5HT parameters. We have shown that animals with constitutively low 5HT activity maintained at a standard housing temperature (22 °C) have greater interscapular BAT (iBAT) mass and higher iBAT metabolic activity (as evidenced by measurements of iBAT temperature and glucose uptake), accompanied by increased iBAT mRNA expression of key thermogenic genes, compared to animals with high 5HT tone. In response to further thermogenic challenges-intermittent cold exposure or treatment with a β3-adrenergic agonist-5HT sublines show several functional and molecular differences linking constitutively low endogenous 5HT tone to higher BAT activity/capacity. Overall, the results support a role of 5-HT in the control of BAT thermogenesis They also suggest that individuals with lower 5HT activity may be more sensitive to β3-adrenergic drugs.
Collapse
Affiliation(s)
- Maja Kesić
- Department of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Petra Baković
- Department of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Vladimir Farkaš
- Department of Experimental Physics, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Robert Bagarić
- Department of Experimental Physics, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Darko Kolarić
- Centre for Informatics and Computing, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Jasminka Štefulj
- Department of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Lipa Čičin-Šain
- Department of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| |
Collapse
|
38
|
Zhou Z, Zhang H, Tao Y, Jie H, Zhao J, Zang J, Li H, Wang Y, Wang T, Zhao H, Li Y, Guo C, Zhu F, Mao H, Zhang L, Liu F, Wang Q. CX3CR1 hi macrophages sustain metabolic adaptation by relieving adipose-derived stem cell senescence in visceral adipose tissue. Cell Rep 2023; 42:112424. [PMID: 37086405 DOI: 10.1016/j.celrep.2023.112424] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/20/2022] [Accepted: 04/06/2023] [Indexed: 04/23/2023] Open
Abstract
Adipose-derived stem cells (ASCs) drive healthy visceral adipose tissue (VAT) expansion via adipocyte hyperplasia. Obesity induces ASC senescence that causes VAT dysfunction and metabolic disorders. It is challenging to restrain this process by biological intervention, as mechanisms of controlling VAT ASC senescence remain unclear. We demonstrate that a population of CX3CR1hi macrophages is maintained in mouse VAT during short-term energy surplus, which sustains ASCs by restraining their senescence, driving adaptive VAT expansion and metabolic health. Long-term overnutrition induces diminishment of CX3CR1hi macrophages in mouse VAT accompanied by ASC senescence and exhaustion, while transferring CX3CR1hi macrophages restores ASC reservoir and triggers VAT beiging to alleviate the metabolic maladaptation. Mechanistically, visceral ASCs attract macrophages via MCP-1 and shape their CX3CR1hi phenotype via exosomes; these macrophages relieve ASC senescence by promoting the arginase1-eIF5A hypusination axis. These findings identify VAT CX3CR1hi macrophages as ASC supporters and unravel their therapeutic potential for metabolic maladaptation to obesity.
Collapse
Affiliation(s)
- Zixin Zhou
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Huiying Zhang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yan Tao
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Haipeng Jie
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jingyuan Zhao
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jinhao Zang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Huijie Li
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yalin Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tianci Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hui Zhao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Yuan Li
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Chun Guo
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Faliang Zhu
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Haiting Mao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Lining Zhang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Fengming Liu
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
39
|
Ye J, Gao C, Liang Y, Hou Z, Shi Y, Wang Y. Characteristic and fate determination of adipose precursors during adipose tissue remodeling. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:13. [PMID: 37138165 PMCID: PMC10156890 DOI: 10.1186/s13619-023-00157-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 12/30/2022] [Indexed: 05/05/2023]
Abstract
Adipose tissues are essential for actively regulating systemic energy balance, glucose homeostasis, immune responses, reproduction, and longevity. Adipocytes maintain dynamic metabolic needs and possess heterogeneity in energy storage and supply. Overexpansion of adipose tissue, especially the visceral type, is a high risk for diabetes and other metabolic diseases. Changes in adipocytes, hypertrophy or hyperplasia, contribute to the remodeling of obese adipose tissues, accompanied by abundant immune cell accumulation, decreased angiogenesis, and aberrant extracellular matrix deposition. The process and mechanism of adipogenesis are well known, however, adipose precursors and their fate decision are only being defined with recent information available to decipher how adipose tissues generate, maintain, and remodel. Here, we discuss the key findings that identify adipose precursors phenotypically, with special emphasis on the intrinsic and extrinsic signals in instructing and regulating the fate of adipose precursors under pathophysiological conditions. We hope that the information in this review lead to novel therapeutic strategies to combat obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Jiayin Ye
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Cheng Gao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Yong Liang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Zongliu Hou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, 650000, Yunnan, China
| | - Yufang Shi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
- The Third Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China.
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| |
Collapse
|
40
|
Colangeli L, Escobar Marcillo DI, Simonelli V, Iorio E, Rinaldi T, Sbraccia P, Fortini P, Guglielmi V. The Crosstalk between Gut Microbiota and White Adipose Tissue Mitochondria in Obesity. Nutrients 2023; 15:nu15071723. [PMID: 37049562 PMCID: PMC10097238 DOI: 10.3390/nu15071723] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/19/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Adipose tissue (AT) dysregulation is a key process in the pathophysiology of obesity and its cardiometabolic complications, but even if a growing body of evidence has been collected over recent decades, the underlying molecular basis of adiposopathy remains to be fully understood. In this context, mitochondria, the intracellular organelles that orchestrate energy production and undergo highly dynamic adaptive changes in response to changing environments, have emerged as crucial regulators of both white (WAT) and brown adipose tissue (BAT) metabolism and function. Given that the gut microbiota and its metabolites are able to regulate host metabolism, adipogenesis, WAT inflammation, and thermogenesis, we hypothesize that their frequently observed dysregulation in obesity could affect AT metabolism by exerting direct and indirect effects on AT mitochondria. By collecting and revising the current evidence on the connections between gut microbiota and AT mitochondria in obesity, we gained insights into the molecular biology of their hitherto largely unexplored crosstalk, tracing how gut microbiota may regulate AT mitochondrial function.
Collapse
|
41
|
Barthelemy J, Bogard G, Wolowczuk I. Beyond energy balance regulation: The underestimated role of adipose tissues in host defense against pathogens. Front Immunol 2023; 14:1083191. [PMID: 36936928 PMCID: PMC10019896 DOI: 10.3389/fimmu.2023.1083191] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/09/2023] [Indexed: 03/06/2023] Open
Abstract
Although the adipose tissue (AT) is a central metabolic organ in the regulation of whole-body energy homeostasis, it is also an important endocrine and immunological organ. As an endocrine organ, AT secretes a variety of bioactive peptides known as adipokines - some of which have inflammatory and immunoregulatory properties. As an immunological organ, AT contains a broad spectrum of innate and adaptive immune cells that have mostly been studied in the context of obesity. However, overwhelming evidence supports the notion that AT is a genuine immunological effector site, which contains all cell subsets required to induce and generate specific and effective immune responses against pathogens. Indeed, AT was reported to be an immune reservoir in the host's response to infection, and a site of parasitic, bacterial and viral infections. In addition, besides AT's immune cells, preadipocytes and adipocytes were shown to express innate immune receptors, and adipocytes were reported as antigen-presenting cells to regulate T-cell-mediated adaptive immunity. Here we review the current knowledge on the role of AT and AT's immune system in host defense against pathogens. First, we will summarize the main characteristics of AT: type, distribution, function, and extraordinary plasticity. Second, we will describe the intimate contact AT has with lymph nodes and vessels, and AT immune cell composition. Finally, we will present a comprehensive and up-to-date overview of the current research on the contribution of AT to host defense against pathogens, including the respiratory viruses influenza and SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Isabelle Wolowczuk
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019 - UMR 9017 - Center for Infection and Immunity of Lille (CIIL), Lille, France
| |
Collapse
|
42
|
Latteri S, Sofia M, Puleo S, Di Vincenzo A, Cinti S, Castorina S. Mechanisms linking bariatric surgery to adipose tissue, glucose metabolism, fatty liver disease and gut microbiota. Langenbecks Arch Surg 2023; 408:101. [PMID: 36826628 PMCID: PMC9957865 DOI: 10.1007/s00423-023-02821-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 01/25/2023] [Indexed: 02/25/2023]
Abstract
PURPOSE In the last 20 years, bariatric surgery has achieved an important role in translational and clinical research because of obesity comorbidities. Initially, a tool to lose weight, bariatric surgery now has been shown to be involved in several metabolic pathways. METHODS We conducted a narrative review discussing the underlying mechanisms that could explain the impact of bariatric surgery and the relationship between obesity and adipose tissue, T2D, gut microbiota, and NAFLD. RESULTS Bariatric surgery has an impact in the relation between obesity and type 2 diabetes, but in addition it induces the white-to-brown adipocyte trans-differentiation, by enhancing thermogenesis. Another issue is the connection of bariatric surgery with the gut microbiota and its role in the complex mechanism underlying weight gain. CONCLUSION Bariatric surgery modifies gut microbiota, and these modifications influence lipid metabolism, leading to improvement of non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Saverio Latteri
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Maria Sofia
- Department of General Surgery, Cannizzaro Hospital, Via Messina 829, 95126, Catania, Italy.
| | - Stefano Puleo
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| | - Angelica Di Vincenzo
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto 10A, 60020, Ancona, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto 10A, 60020, Ancona, Italy
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| |
Collapse
|
43
|
Song Z, Liu N, He Y, Chen J, Li J, Wang F, Wu Z. Knockout of ICAT in Adipose Tissue Alleviates Fibro-inflammation in Obese Mice. Inflammation 2023; 46:404-417. [PMID: 36181623 DOI: 10.1007/s10753-022-01742-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022]
Abstract
The E2 promoter binding factor 1 (E2F1) and the Wnt/β-catenin signaling are crucial in regulating metabolic homeostasis including obesity. The β-catenin interacting protein 1 (CTNNBIP1), also known as the inhibitor of β-catenin and TCF4 (ICAT), is required for E2F1 to inhibit the activity of β-catenin. However, the role of ICAT in E2F1 regulating obesity-related metabolic disorders remains unknown. In the present study, male adipose tissue-specific ICAT knockout (ICATadi-/-) C57BL/6 J mice and control littermates aged 6-8 weeks were fed with high-fat diet (HFD) for 12 weeks to explore the effect of ICAT on lipid metabolism and obesity-related disorders. Results showed that the adipose tissue-specific ICAT knockout had negligible effect on lipid metabolism, reflected by no difference in body weight, fat mass, and the expression of proteins involved in lipid metabolism in white adipose tissue (WAT) and the liver between the ICATadi-/- mice and the control littermate (ICATfl/fl) mice. However, the knockout of ICAT reduced inflammatory response in WAT and the liver. Additionally, Sirius red staining results showed that deletion of ICAT attenuated fibrosis and reduced mRNA levels of transforming growth factor β1(TGF-β1), matrix metallopeptidase 2 (Mmp2), Mmp3, and collagen, type V, alpha 1 (Col5a1) in WAT and the liver. These results suggested that knockout of ICAT improved the metabolic abnormalities of obese mice through attenuating adipose tissue and the liver inflammation as well as fibrosis. Our findings may provide a new insight to understand the role of ICAT in inflammation and fibrosis.
Collapse
Affiliation(s)
- Zhuan Song
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Ning Liu
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China
| | - Yu He
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Jingqing Chen
- Laboratory Animal Center of the Academy of Military Medical Sciences, Beijing, 100193, China
| | - Jun Li
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Fengchao Wang
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Zhenlong Wu
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China. .,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
44
|
Tumor and peritumoral adipose tissue crosstalk: De-differentiated adipocytes influence spread of colon carcinoma cells. Tissue Cell 2023; 80:101990. [PMID: 36542947 DOI: 10.1016/j.tice.2022.101990] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022]
Abstract
Colorectal cancer is the second leading cause of cancer and often has a fatal course. There are many studies in the literature that have described a close functional relationship between the tumor mass and surrounding tissue, or tumor stroma, which is affected by the continuous metabolic exchange that occurs at the interface between tumor and tissues in contact with it. There is much evidence that the presence of adipose tissue in stroma plays a fundamental role in modulating the tumor microenvironment and promote tumor development, growth, and angiogenesis due to its endocrine characteristics. In this analysis, we have studied the alterations of adipose tissue surrounding colorectal tumors with MRI and optical imaging in vivo techniques to monitor tumor progression and also performed histological and molecular analysis. We detected differences in the principal adipose markers expressed by adipocytes residing around the rectal colon and observed that peritumoral adipose tissue is exposed to a mesenchymal transition process that leads to the acquisition of a less differentiated phenotype of adipocyte that represents the main cellular type present in tumor stroma. The mesenchymal transition correlated with the acquisition of more aggressive tumor phenotype and could represent a valid target for tumor therapy.
Collapse
|
45
|
Vliora M, Ravelli C, Grillo E, Corsini M, Flouris AD, Mitola S. The impact of adipokines on vascular networks in adipose tissue. Cytokine Growth Factor Rev 2023; 69:61-72. [PMID: 35953434 DOI: 10.1016/j.cytogfr.2022.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 02/07/2023]
Abstract
Adipose tissue (AT) is a highly active and plastic endocrine organ. It secretes numerous soluble molecules known as adipokines, which act locally to AT control the remodel and homeostasis or exert pleiotropic functions in different peripheral organs. Aberrant production or loss of certain adipokines contributes to AT dysfunction associated with metabolic disorders, including obesity. The AT plasticity is strictly related to tissue vascularization. Angiogenesis supports the AT expansion, while regression of blood vessels is associated with AT hypoxia, which in turn mediates tissue inflammation, fibrosis and metabolic dysfunction. Several adipokines can regulate endothelial cell functions and are endowed with either pro- or anti-angiogenic properties. Here we address the role of adipokines in the regulation of angiogenesis. A better understanding of the link between adipokines and angiogenesis will open the way for novel therapeutic approaches to treat obesity and metabolic diseases.
Collapse
Affiliation(s)
- Maria Vliora
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece; Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Andreas D Flouris
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy.
| |
Collapse
|
46
|
Grillo E, Ravelli C, Colleluori G, D'Agostino F, Domenichini M, Giordano A, Mitola S. Role of gremlin-1 in the pathophysiology of the adipose tissues. Cytokine Growth Factor Rev 2023; 69:51-60. [PMID: 36155165 DOI: 10.1016/j.cytogfr.2022.09.004] [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: 08/12/2022] [Accepted: 09/12/2022] [Indexed: 02/07/2023]
Abstract
Gremlin-1 is a secreted bone morphogenetic protein (BMP) antagonist playing a pivotal role in the regulation of tissue formation and embryonic development. Since its first identification in 1997, gremlin-1 has been shown to be a multifunctional factor involved in wound healing, inflammation, cancer and tissue fibrosis. Among others, the activity of gremlin-1 is mediated by its interaction with BMPs or with membrane receptors such as the vascular endothelial growth factor receptor 2 (VEGFR2) or heparan sulfate proteoglycans (HSPGs). Growing evidence has highlighted a central role of gremlin-1 in the homeostasis of the adipose tissue (AT). Of note, gremlin-1 is involved in AT dysfunction during type 2 diabetes, obesity and non-alcoholic fatty liver disease (NAFLD) metabolic disorders. In this review we discuss recent findings on gremlin-1 involvement in AT biology, with particular attention to its role in metabolic diseases, to highlight its potential as a prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Georgia Colleluori
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Francesco D'Agostino
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mattia Domenichini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
47
|
Effects of Fatty Acid Metabolites on Adipocytes Britening: Role of Thromboxane A2. Cells 2023; 12:cells12030446. [PMID: 36766790 PMCID: PMC9913700 DOI: 10.3390/cells12030446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Obesity is a complex disease highly related to diet and lifestyle and is associated with low amount of thermogenic adipocytes. Therapeutics that regulate brown adipocyte recruitment and activity represent interesting strategies to fight overweight and associated comorbidities. Recent studies suggest a role for several fatty acids and their metabolites, called lipokines, in the control of thermogenesis. The purpose of this work was to analyze the role of several lipokines in the control of brown/brite adipocyte formation. We used a validated human adipocyte model, human multipotent adipose-derived stem cell model (hMADS). In the absence of rosiglitazone, hMADS cells differentiate into white adipocytes, but convert into brite adipocytes upon rosiglitazone or prostacyclin 2 (PGI2) treatment. Gene expression was quantified using RT-qPCR and protein levels were assessed by Western blotting. We show here that lipokines such as 12,13-diHOME, 12-HEPE, 15dPGJ2 and 15dPGJ3 were not able to induce browning of white hMADS adipocytes. However, both fatty acid esters of hydroxy fatty acids (FAHFAs), 9-PAHPA and 9-PAHSA potentiated brown key marker UCP1 mRNA levels. Interestingly, CTA2, the stable analog of thromboxane A2 (TXA2), but not its inactive metabolite TXB2, inhibited the rosiglitazone and PGI2-induced browning of hMADS adipocytes. These results pinpoint TXA2 as a lipokine inhibiting brown adipocyte formation that is antagonized by PGI2. Our data open new horizons in the development of potential therapies based on the control of thromboxane A2/prostacyclin balance to combat obesity and associated metabolic disorders.
Collapse
|
48
|
Salvio G, Petrelli M, Paolini S, Baldini V, Sbaffi C, Basili S, Giordano A, Balercia G, Cinti S. Gender-specific effects of capsiate supplementation on body weight and bone mineral density: a randomized, double-blind, placebo-controlled study in slightly overweight women. J Endocrinol Invest 2023:10.1007/s40618-022-01999-w. [PMID: 36609773 DOI: 10.1007/s40618-022-01999-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Overweight and obesity are highly prevalent conditions associated with premature morbidity and mortality worldwide. Capsiate, a nonpungent analogue of capsaicin, binds to TRP vanilloid 1 (TRPV1) receptor, which is involved in adipogenesis, and could be effective as a weight-lowering agent. METHODS Eighteen slightly overweight women were enrolled in this randomized, double-blind, placebo-controlled study. Nine patients were included in the capsiate intervention group and received 9 mg/day of capsinoids and 9 patients received placebo for 8 weeks. All patients underwent weight and waist circumference assessment before and after treatment. Body composition and bone mineral density (BMD) were also detected by dual-energy X-ray absorptiometry (DXA). RESULTS Fourteen patients completed the study. The treatment with capsiate or placebo for 8 weeks was not associated with significant changes in weight or waist circumference. After treatment, there was a significant improvement in BMD values measured at the spine in the capsiate group (1.158 vs 1.106 g/cm2, + 4.7%; p = 0.04), but not in the group treated with placebo. Similarly, the capsiate group showed a 9.1% increase (p = 0.05) in the adipose tissue and an 8.5% decrease in lean mass measured at the supraclavicular level, whereas these changes were not statistically significant in the placebo group. CONCLUSIONS Treatment with capsiate for 8 weeks led to negligible changes in body weight in a small sample of slightly overweight women, but our findings suggest a potential effect of capsaicin on bone metabolism in humans.
Collapse
Affiliation(s)
- G Salvio
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - M Petrelli
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - S Paolini
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - V Baldini
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - C Sbaffi
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - S Basili
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - A Giordano
- Center of Obesity, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - G Balercia
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - S Cinti
- Center of Obesity, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy.
| |
Collapse
|
49
|
Migliolo L, de A. Boleti A, de O. Cardoso P, Frihling BF, e Silva P, de Moraes LRN. Adipose tissue, systematic inflammation, and neurodegenerative diseases. Neural Regen Res 2023; 18:38-46. [PMID: 35799506 PMCID: PMC9241402 DOI: 10.4103/1673-5374.343891] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
50
|
Tozzi R, Campolo F, Baldini E, Venneri MA, Lubrano C, Ulisse S, Gnessi L, Mariani S. Ketogenic Diet Increases Serum and White Adipose Tissue SIRT1 Expression in Mice. Int J Mol Sci 2022; 23:ijms232415860. [PMID: 36555502 PMCID: PMC9785229 DOI: 10.3390/ijms232415860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Overnutrition and its sequelae have become a global concern due to the increasing incidence of obesity and insulin resistance. A ketogenic diet (KD) is widely used as a dietary treatment for metabolic disorders. Sirtuin1 (SIRT1), a metabolic sensor which regulates fat homeostasis, is modulated by dietary interventions. However, the influence of nutritional ketosis on SIRT1 is still debated. We examined the effect of KD on adipose tissue, liver, and serum levels of SIRT1 in mice. Adult C57BL/6J male mice were randomly assigned to two isocaloric dietary groups and fed with either high-fat KD or normal chow (NC) for 4 weeks. Serum SIRT1, beta-hydroxybutyrate (βHB), glucose, and triglyceride levels, as well as SIRT1 expression in visceral (VAT), subcutaneous (SAT), and brown (BAT) adipose tissues, and in the liver, were measured. KD-fed mice showed an increase in serum βHB in parallel with serum SIRT1 (r = 0.732, p = 0.0156), and increased SIRT1 protein expression in SAT and VAT. SIRT1 levels remained unchanged in BAT and in the liver, which developed steatosis. Normal glycemia and triglycerides were observed. Under a KD, serum and white fat phenotypes show higher SIRT1, suggesting that one of the molecular mechanisms underlying a KD's potential benefits on metabolic health involves a synergistic interaction with SIRT1.
Collapse
Affiliation(s)
- Rossella Tozzi
- Department of Molecular Medicine, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Enke Baldini
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Carla Lubrano
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Salvatore Ulisse
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Lucio Gnessi
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Stefania Mariani
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
- Correspondence: ; Tel.: +39-6-49970509; Fax: +39-6-4461450
| |
Collapse
|