1
|
Ding L, Lu Z, Jiang X, Zhang S, Tian X, Wang Q. Obesity-derived macrophages upregulate TNF-α to induce apoptosis in glial cell via the NF-κB/PHLPP1 axis. Int Immunopharmacol 2024; 141:112962. [PMID: 39197294 DOI: 10.1016/j.intimp.2024.112962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024]
Abstract
Macrophages in obese adipose tissue have been shown to damage nerve fibers, however, the mechanism underlying how macrophages cause glial cell damage remains unknown. This study aimed to characterize the mechanism by which macrophages induce apoptosis in glial cell during obesity formation in mice by single-nucleus RNA sequencing (snRNA-seq). Cells obtained from paraepididymal adipose tissue in obese mice underwent snRNA-seq. Eighteen different clusters were identified, and 12 cell types were annotated, including glial cells, macrophages, and fibroblasts. There was a negative correlation between the number of glial cells and macrophages in mouse adipose tissue during the formation of obesity. The pro-apoptotic factor PHLPP1 was identified in GO Terms. The interaction between adipose tissue glial cells and macrophages was revealed via in-depth analysis, and the cell-cell communication mechanism between the TNF-α and NF-KB/PHLPP1 axes was perfected. Apoptosis of glial cell by upregulation of TNF-α via obesity-derived macrophages and activation of the NF-κB/PHLPP1 axis. We further revealed how macrophages induce apoptosis in glial cells during obesity formation, as well as different changes in the two cell populations. This study provides valuable resources and foundations for understanding the mechanistic effects of macrophages and glial cells during obesity formation, as well as diseases and potential interventions.
Collapse
Affiliation(s)
- Ling Ding
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Zhimin Lu
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Xing Jiang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Sen Zhang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Xuewen Tian
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China.
| | - Qinglu Wang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China.
| |
Collapse
|
2
|
Jiang T, Zou A, Song W, Zheng J, Lai L, Wang Q, Cui B. Interleukin-27 signaling resists obesity by promoting the accumulation of Treg cells in visceral adipose tissue. Biochem Biophys Res Commun 2024; 733:150690. [PMID: 39276693 DOI: 10.1016/j.bbrc.2024.150690] [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: 08/31/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
The prevalence of obesity and its associated metabolic disorders has emerged as one of the most significant health threats worldwide. The visceral adipose tissue regulatory T cells (VAT Treg) play an essential role in maintaining homeostasis and preventing obesity mainly by secreting Interleikin-10 (IL-10) and Transforming Growth Factor β (TGF-β). However, the mechanism that regulates VAT Treg quantity and function remains unclear. Here we elucidate the pivotal role of IL-27 signaling in sustaining the accumulation of VAT Treg cells, thereby conferring protection against obesity. We found that mice with the deficiency of IL-27 receptor Wsx1 gained more body weight and VAT weight than their wild-type littermates when fed both a normal-fat diet (NFD) and a high-fat diet (HFD). Notably, the population of VAT Treg cells was reduced in Wsx1 knockout (KO) mice, regardless of whether they were fed a normal-fat diet (NFD) or a high-fat diet (HFD). Correspondingly, the expression levels of the transcription factors FOXP3 and PPAR-γ, essential for VAT Treg function, were also diminished in Wsx1 KO mice. Taken together, our findings indicate that IL-27 signaling plays a protective role in obesity by supporting the maintenance and accumulation of VAT Treg cells.
Collapse
Affiliation(s)
- Tianqi Jiang
- Laboratory Medicine Center, Zhejiang Center for Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, PR China
| | - Aixuan Zou
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Wenjun Song
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Jialing Zheng
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Lihua Lai
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China.
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China.
| | - Bijun Cui
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, PR China; Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China.
| |
Collapse
|
3
|
Quarta S, Santarpino G, Carluccio MA, Calabriso N, Cardetta F, Siracusa L, Strano T, Palamà I, Leccese G, Visioli F, Massaro M. Cardiac fat adipocytes: An optimized protocol for isolation of ready-to-use mature adipocytes from human pericardial adipose tissue. J Mol Cell Cardiol 2024; 196:12-25. [PMID: 39214497 DOI: 10.1016/j.yjmcc.2024.08.006] [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: 05/03/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
A better understanding of the pathophysiology of cardiac fat depots is crucial to describe their role in the development of cardiovascular diseases. To this end, we have developed a method to isolate mature fat cells from the pericardial adipose tissue (PAT), the most accessible cardiac fat depot during cardiac surgery. Using enzymatic isolation, we were able to successfully obtain mature fat cells together with the corresponding cells of the stromal vascular fraction (SVF). We subjected the PAT adipocytes to thorough morphological and molecular characterization, including detailed fatty acid profiling, and simultaneously investigated their reactivity to external stimuli. Our approach resulted in highly purified fat cells with sustained viability for up to 72 h after explantation. Remarkably, these adipocytes responded to multiple challenges, including pro-inflammatory and metabolic stimuli, indicating their potential to trigger a pro-inflammatory response and modulate endothelial cell behavior. Furthermore, we have created conditions to maintain whole PAT in culture and preserve their viability and reactivity to external stimuli. The efficiency of cell recovery combined with minimal dedifferentiation underscores the promise for future applications as a personalized tool for screening and assessing individual patient responses to drugs and supplements or nutraceuticals.
Collapse
Affiliation(s)
- Stefano Quarta
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy.
| | - Giuseppe Santarpino
- Department of Clinical and Experimental Medicine, Magna Graecia University of Catanzaro, Italy; Department of Cardiac Surgery, Città di Lecce Hospital, GVM Care&Research, Lecce, Italy; Department of Cardiac Surgery, Paracelsus Medical University, Nuremberg, Germany.
| | | | - Nadia Calabriso
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy.
| | - Francesco Cardetta
- Department of Cardiac Surgery, University "Campus Biomedico", Rome, Italy.
| | - Laura Siracusa
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Catania section, Via Paolo Gaifami 18, 95126 Catania, Italy.
| | - Tonia Strano
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Catania section, Via Paolo Gaifami 18, 95126 Catania, Italy.
| | - Ilaria Palamà
- Institute Nanotechnology Institute, CNR-NANOTEC, 73100 Lecce, Italy.
| | - Gabriella Leccese
- Institute Nanotechnology Institute, CNR-NANOTEC, 73100 Lecce, Italy.
| | | | - Marika Massaro
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy.
| |
Collapse
|
4
|
Xiao L, Mochizuki M, Shimamura N, Sunada K, Nakahara T. Interplay of co-cultured chimeric adipose and gingival tissues exacerbates inflammatory dysfunction relevant to periodontal and metabolic conditions. Life Sci 2024; 355:123009. [PMID: 39197574 DOI: 10.1016/j.lfs.2024.123009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024]
Abstract
Adipose tissue dysfunction is a key feature of metabolic syndrome, which increases the risk of periodontitis, an inflammatory disease induced by bacteria that affects the gingiva and other components of periodontal tissue. Recent studies indicate that molecules from inflamed periodontal tissue contribute to adipose tissue dysfunction. However, the cellular mechanisms and interactions between adipose tissue and gingiva driving the progression of metabolic and periodontal conditions remain unclear. To address this, we developed a chimeric (mouse/human) co-culture tissue model (which identifies the origins of species-specific cytokines) to investigate these interactions. Using tissue-specific functional cells and immunocytes, we constructed equivalents of adipose tissue (ATE) and gingiva (GTE), co-cultivating them under inflammatory conditions induced by bacterial endotoxin, lipopolysaccharide (LPS). Our findings showed that exposure to LPS resulted in a notable reduction in lipid accumulation, GLUT4 expression, and adiponectin secretion in ATE, along with increased macrophage colonies forming around lipid droplets, as well as elevated levels of triglyceride, leptin, and IL-6. In GTE, LPS triggered significant inflammatory responses, characterized by increased macrophage accumulation, elevated COX-2 expression, and heightened secretion of inflammatory cytokines. LPS also reduced epithelial thickness and the expression of keratin 19 and collagen IV, indicating impaired barrier function and gingival integrity. Co-culturing ATE with GTE exacerbated these LPS-induced harmful effects in both tissues. In conclusion, our findings suggest that interplay between gingiva and adipose tissue can intensify the inflammatory and dysfunctional changes caused by LPS. This co-culture tissue model offers a valuable tool for future studies on periodontitis and metabolic syndrome.
Collapse
Affiliation(s)
- Li Xiao
- Department of Physiology, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo 102-8159, Japan.
| | - Mai Mochizuki
- Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, Japan; Department of Life Science Dentistry, The Nippon Dental University, Japan.
| | - Naohiro Shimamura
- Department of Dental Anesthesiology, School of Life Dentistry at Tokyo, The Nippon Dental University, Japan.
| | - Katsuhisa Sunada
- Department of Dental Anesthesiology, School of Life Dentistry at Tokyo, The Nippon Dental University, Japan.
| | - Taka Nakahara
- Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, Japan.
| |
Collapse
|
5
|
Hu T, Liu CH, Lei M, Zeng Q, Li L, Tang H, Zhang N. Metabolic regulation of the immune system in health and diseases: mechanisms and interventions. Signal Transduct Target Ther 2024; 9:268. [PMID: 39379377 PMCID: PMC11461632 DOI: 10.1038/s41392-024-01954-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/18/2024] [Accepted: 08/11/2024] [Indexed: 10/10/2024] Open
Abstract
Metabolism, including glycolysis, oxidative phosphorylation, fatty acid oxidation, and other metabolic pathways, impacts the phenotypes and functions of immune cells. The metabolic regulation of the immune system is important in the pathogenesis and progression of numerous diseases, such as cancers, autoimmune diseases and metabolic diseases. The concept of immunometabolism was introduced over a decade ago to elucidate the intricate interplay between metabolism and immunity. The definition of immunometabolism has expanded from chronic low-grade inflammation in metabolic diseases to metabolic reprogramming of immune cells in various diseases. With immunometabolism being proposed and developed, the metabolic regulation of the immune system can be gradually summarized and becomes more and more clearer. In the context of many diseases including cancer, autoimmune diseases, metabolic diseases, and many other disease, metabolic reprogramming occurs in immune cells inducing proinflammatory or anti-inflammatory effects. The phenotypic and functional changes of immune cells caused by metabolic regulation further affect and development of diseases. Based on experimental results, targeting cellular metabolism of immune cells becomes a promising therapy. In this review, we focus on immune cells to introduce their metabolic pathways and metabolic reprogramming, and summarize how these metabolic pathways affect immune effects in the context of diseases. We thoroughly explore targets and treatments based on immunometabolism in existing studies. The challenges of translating experimental results into clinical applications in the field of immunometabolism are also summarized. We believe that a better understanding of immune regulation in health and diseases will improve the management of most diseases.
Collapse
Affiliation(s)
- Tengyue Hu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Chang-Hai Liu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Min Lei
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qingmin Zeng
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Division of Renal and endocrinology, Qin Huang Hospital, Xi'an, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China.
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Nannan Zhang
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China.
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Guo C, He L, Tu Y, Xu C, Liao C, Lai H, Lin C, Tu H. Insulin resistance and sarcopenia: a prognostic longitudinal link to stroke risk in middle-aged and elderly Chinese population. BMC Public Health 2024; 24:2757. [PMID: 39385146 PMCID: PMC11465621 DOI: 10.1186/s12889-024-20214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/27/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Stroke is the leading cause of death in middle-aged and elderly people in China. Insulin resistance (IR) and sarcopenia are both closely associated with metabolic diseases. However, the relationship between these two indicators and stroke has not been fully investigated. The aim of this study was to investigate the relationship between IR and sarcopenia and the risk of new-onset stroke. METHODS Using longitudinal data from the China Health and Retirement Longitudinal Study (CHARLS) from 2011 to 2018, Cox proportional hazards models were used to determine the association between IR surrogate indicators and sarcopenia status with stroke incidence. RESULTS In the present study, during a median 7 years of follow-up, we included 7009 middle-aged and elderly residents, of whom 515 presented with stroke incidence. After adjustment for potential confounders, both baseline IR surrogates and sarcopenia independently predicted stroke risk. In addition, co-morbidities had a higher risk of stroke than other groups. The positive association between TyG-WC and sarcopenia on stroke risk was particularly significant [HR (95% CI): 2.03 (1.52, 2.70)]. In subgroups of different ages and sexes, the combination of IR and sarcopenia is associated with the highest risk of stroke. CONCLUSIONS We found that IR and sarcopenia synergistically increase the incidence of stroke in older adults. This finding provides new perspectives for stroke detection and intervention and highlights the importance of early detection and management of IR and sarcopenia in older adults.
Collapse
Affiliation(s)
- Canhui Guo
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- The Department of Geriatrics, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, Jiangxi, 330006, China
| | - Ling He
- The Department of Geriatrics, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, Jiangxi, 330006, China
| | - Yansong Tu
- Faculty of Science, University of Melbourne Grattan Street, Parkville, VIC, 3010, Australia
| | - Chunyan Xu
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- The Department of Geriatrics, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, Jiangxi, 330006, China
| | - Caifeng Liao
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- The Department of Geriatrics, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, Jiangxi, 330006, China
| | - Hurong Lai
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- The Department of Geriatrics, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, Jiangxi, 330006, China
| | - Chuyang Lin
- The Department of Geriatrics, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, Jiangxi, 330006, China
| | - Huaijun Tu
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- The Department of Geriatrics, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
7
|
Zhang X, Liang J, Luo H, Zhang H, Xiang J, Guo L, Zhu X. The association between body roundness index and osteoporosis in American adults: analysis from NHANES dataset. Front Nutr 2024; 11:1461540. [PMID: 39430785 PMCID: PMC11486732 DOI: 10.3389/fnut.2024.1461540] [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: 08/12/2024] [Accepted: 09/17/2024] [Indexed: 10/22/2024] Open
Abstract
Background An innovative way to quantify obesity that appropriately captures levels of visceral and body fat is the Body Roundness Index (BRI). The purpose of this study is to look at the relationship between BRI and osteoporosis (OP) in adult Americans. Methods This study utilized data from the National Health and Nutrition Examination Survey (NHANES) collected between 2007 and 2018. NHANES is a research program designed to assess the health and nutritional status of adults and children in the United States. It conducts surveys focusing on various populations and health-related topics. Logistic regression analysis was employed to investigate the relationship between BRI and OP, adjusting for various covariates. BRI was categorized into four levels to further explore the association trends between different BRI levels and OP, enhancing the robustness of the results. Using restricted cubic spline (RCS) analysis, the dose-response relationship between BRI and OP was illustrated. Subgroup analyses were also carried out to evaluate the consistency and robustness of the findings. Results This study included 8,899 participants aged 50 years and older, among whom 763 had OP. BRI and the prevalence of OP were inversely correlated in the fully adjusted model (OR = 0.79, 95% CI: 0.69-0.86). The prevalence of OP considerably reduced with higher BRI levels when BRI was converted from a continuous to a categorical variable in comparison to the lowest BRI quartile. RCS analysis revealed an L-shaped negative correlation between BRI and OP prevalence, with a threshold effect analysis identifying a breakpoint at BRI = 5.29. Each unit increase in BRI to the left of this breakpoint was linked to a 36% decrease in the probability of OP (OR = 0.64, 95% CI: 0.57-0.72). Based on stratified factor subgroup analyses, it was shown that the negative correlation between BRI and OP persisted. Conclusion In a large, representative sample of American adults, this study identified a significant negative correlation between BRI and the prevalence of OP. Specifically, as BRI increases, the prevalence of osteoporosis decreases. Maintaining an appropriate and healthy BRI level may play a critical role in the prevention of osteoporosis. Therefore, regular monitoring of BRI and the adoption of appropriate health measures are essential for reducing the risk of osteoporosis.
Collapse
Affiliation(s)
- Xunmeng Zhang
- The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiarong Liang
- The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hong Luo
- Longhu Street Community Health Service Center, Guangzhou, China
| | | | - Jing Xiang
- The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lianjin Guo
- The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuemin Zhu
- The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
8
|
Hill KB, Mullen GP, Nagareddy PR, Zimmerman KA, Rudolph MC. Key questions and gaps in understanding adipose tissue macrophages and early-life metabolic programming. Am J Physiol Endocrinol Metab 2024; 327:E478-E497. [PMID: 39171752 PMCID: PMC11482221 DOI: 10.1152/ajpendo.00140.2024] [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: 04/10/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 08/23/2024]
Abstract
The global obesity epidemic, with its associated comorbidities and increased risk of early mortality, underscores the urgent need for enhancing our understanding of the origins of this complex disease. It is increasingly clear that metabolism is programmed early in life and that metabolic programming can have life-long health consequences. As a critical metabolic organ sensitive to early-life stimuli, proper development of adipose tissue (AT) is crucial for life-long energy homeostasis. Early-life nutrients, especially fatty acids (FAs), significantly influence the programming of AT and shape its function and metabolism. Of growing interest are the dynamic responses during pre- and postnatal development to proinflammatory omega-6 (n6) and anti-inflammatory omega-3 (n3) FA exposures in AT. In the US maternal diet, the ratio of "pro-inflammatory" n6- to "anti-inflammatory" n3-FAs has grown dramatically due to the greater prevalence of n6-FAs. Notably, AT macrophages (ATMs) form a significant population within adipose stromal cells, playing not only an instrumental role in AT formation and maintenance but also acting as key mediators of cell-to-cell lipid and cytokine signaling. Despite rapid advances in ATM and immunometabolism fields, research has focused on responses to obesogenic diets and during adulthood. Consequently, there is a significant gap in identifying the mechanisms contributing metabolic health, especially regarding lipid exposures during the establishment of ATM physiology. Our review highlights the current understanding of ATM diversity, their critical role in AT, their potential role in early-life metabolic programming, and the broader implications for metabolism and health.
Collapse
Affiliation(s)
- Kaitlyn B Hill
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Gregory P Mullen
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Cardiovascular Section, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Kurt A Zimmerman
- Department of Internal Medicine, Division of Nephrology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Michael C Rudolph
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
9
|
Park DJ, Choi W, Sayeed S, Dorschner RA, Rainaldi J, Ho K, Kezios J, Nolan JP, Mali P, Costantini T, Eliceiri BP. Defining the activity of pro-reparative extracellular vesicles in wound healing based on miRNA payloads and cell type-specific lineage mapping. Mol Ther 2024; 32:3059-3079. [PMID: 38379282 PMCID: PMC11403212 DOI: 10.1016/j.ymthe.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/02/2024] [Accepted: 02/15/2024] [Indexed: 02/22/2024] Open
Abstract
Small extracellular vesicles (EVs) are released by cells and deliver biologically active payloads to coordinate the response of multiple cell types in cutaneous wound healing. Here we used a cutaneous injury model as a donor of pro-reparative EVs to treat recipient diabetic obese mice, a model of impaired wound healing. We established a functional screen for microRNAs (miRNAs) that increased the pro-reparative activity of EVs and identified a down-regulation of miR-425-5p in EVs in vivo and in vitro associated with the regulation of adiponectin. We tested a cell type-specific reporter of a tetraspanin CD9 fusion with GFP to lineage map the release of EVs from macrophages in the wound bed, based on the expression of miR-425-5p in macrophage-derived EVs and the abundance of macrophages in EV donor sites. Analysis of different promoters demonstrated that EV release under the control of a macrophage-specific promoter was most abundant and that these EVs were internalized by dermal fibroblasts. These findings suggested that pro-reparative EVs deliver miRNAs, such as miR-425-5p, that stimulate the expression of adiponectin that has insulin-sensitizing properties. We propose that EVs promote intercellular signaling between cell layers in the skin to resolve inflammation, induce proliferation of basal keratinocytes, and accelerate wound closure.
Collapse
Affiliation(s)
- Dong Jun Park
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA
| | - Wooil Choi
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA
| | - Sakeef Sayeed
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA
| | - Robert A Dorschner
- Department of Dermatology, University of California San Diego, La Jolla, CA 92093, USA
| | - Joseph Rainaldi
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Kayla Ho
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA
| | - Jenny Kezios
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA
| | | | - Prashant Mali
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Todd Costantini
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA
| | - Brian P Eliceiri
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; Department of Dermatology, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
10
|
Zhao C, Wen Z, Gao Y, Xiao F, Yan J, Wang X, Meng T. Pantothenic Acid Alleviates Fat Deposition and Inflammation by Suppressing the JNK/P38 MAPK Signaling Pathway. J Med Food 2024; 27:834-843. [PMID: 38949913 DOI: 10.1089/jmf.2023.k.0292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024] Open
Abstract
Excessive fat deposition leads to obesity and cardiovascular diseases with abnormal metabolism. Pantothenic acid (PA) is a major B vitamin required for energy metabolism. However, the effect of PA on lipid metabolism and obesity has not been explored. We investigated the effects and molecular mechanism of PA on fat accumulation as well as the influence of adipogenic marker genes in both adult male mice and primary adipocytes. First, we demonstrated that PA attenuates weight gain in mice fed high-fat diet (HFD). Besides, PA supplementation substantially improved glucose tolerance and lipid metabolic disorder in obese mice. Furthermore, PA significantly inhibited white adipose tissue (WAT) deposition as well as fat droplets visualized by magnification in both chow and HFD group. More importantly, PA obviously suppressed the mRNA levels of CD36, IL-6, and TNF-α to alleviate inflammation and reduced the levels of PPARγ, aP2, and C/EBPα genes that are related to lipid metabolism in inguinal white adipose tissue (ing-WAT) and epididymal white adipose tissue (ei-WAT). In vitro, PA supplementation showed a lower lipid droplet aggregation as well as reduced expression levels of adipogentic genes. Finally, we identified that PA inhibits the phosphorylation levels of p38 and JNK in murine primary adipocytes. Collectively, our data demonstrated for the first time that PA attenuates lipid metabolic disorder as well as fat deposition by JNK/p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Cunzhen Zhao
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Ziwei Wen
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Yunfei Gao
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Fang Xiao
- Pingqiao District Bureau of Agriculture and Rural Development of Xinyang, Xinyang, China
| | - Jinzhao Yan
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Xiaotong Wang
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Tiantian Meng
- College of Life Science, Xinyang Normal University, Xinyang, China
| |
Collapse
|
11
|
Saglam E, Karagedik H, Dinc M, Oke D, Gun Atak P, Karadeniz B, Burul G, Gormus Degrigo U. Can Bone Morphogenetic Protein 1 (BMP1) Be a Potential Biomarker of Obesity? Cureus 2024; 16:e67025. [PMID: 39280566 PMCID: PMC11402472 DOI: 10.7759/cureus.67025] [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] [Accepted: 05/07/2024] [Indexed: 09/18/2024] Open
Abstract
Background Obesity has long been a severe threat to public health as an epidemic, and studies on its pathogenesis and treatment have been ongoing. Our study aims to compare the serum levels of bone morphogenetic protein 1 (BMP1), neuregulin 4 (NRG4), and apolipoprotein A5 (ApoA5) in obese and non-obese individuals and investigate their association with obesity. Methodology Our study included a total of 111 participants, of whom 46 were obese (body mass index (BMI) ≥30 kg/m2), aged 18-65 years, and had no comorbidities, and 65 were non-obese (BMI = 18.5-29.9 kg/m2) without any additional disease. For all participants, BMP1, NRG4, and ApoA5 levels were determined and compared with clinical and biochemical parameters. Results Overall, 60.4% (n = 67) of the participants were female and 39.6% (n = 44) were male. In terms of the BMI scores, 58.6% (n = 65) had a BMI <30 kg/m2 and 41.4% (n = 46) had a BMI ≥30 kg/m2. Both, the BMI and the gender groups did not differ significantly in terms of age (p = 0.093 and p = 0.795, respectively). The weight, fat-free mass, mineral quantity, protein quantity, fluid weight, and fluid ratio values of the male participants were significantly higher than females (p = 0.011, p = 0.001, p = 0.001, p = 0.001, p = 0.001, and p = 0.001, respectively). The aspartate aminotransferase (AST)/alanine aminotransferase (ALT) ratios and the triglyceride/glucose (TG/Glu) ratios were found to be significantly higher in males than in females (p = 0.001 and p = 0.001, respectively). The respective BMP1 (15.88 vs. 13.35), AST/ALT (1.36 vs. 1.04) and TG/Glu ratios (1.47 vs. 1.29) were significantly higher, while the quantitative insulin sensitivity check index (QUICKI) was lower in obese individuals than in non-obese individuals (0.32 vs. 0.34). NRG4 and ApoA5 values were similar between the two groups. BMP1, QUICKI values, and AST/ALT ratios proved to be statistically significant in obesity through the univariable logistic regression analysis (β = 1.066, p = 0.048; β = 0.0001, p = 0.001, and β = 3.707, p = 0.003, respectively). On multiple logistic regression analysis, QUICKI values (β = 0.001, p = 0.001) had a negative and significant effect on obesity, and the AST/ALT ratios (β = 2.803, p = 0.033) had a positive and significant effect on obesity. Conclusions Our study indicates that detecting an important link between BMP1 in obese patients will help elucidate the pathogenesis of obesity and come up with a potential therapeutic candidate. BMP1 levels, along with AST/ALT and TG/Glu ratios, were significantly higher in obese patients. BMP1 levels were also an independent significant predictor of obesity together with AST/ALT ratio and QUICKI in this study, suggesting that it may exhibit a metabolic deterioration in obese individuals. However, the results cannot absolutely tell whether it supported deterioration or was a component of the repair mechanism. Althoughit is generally known from recent studies that BMP1 plays a role in osteogenesis, some encouraging results were obtained in our study indicating that BMP1 may play a role in the pathogenesis of obesity. It is expected that our results will not only promote the elucidation of the pathogenesis of obesity, but also provide a therapeutic agent.
Collapse
Affiliation(s)
- Emel Saglam
- Internal Medicine, Bagcilar Training and Research Hospital, Istanbul, TUR
| | - Hande Karagedik
- Molecular Medicine, Aziz Sancar Institute for Experimental Medicine, Istanbul, TUR
| | - Mustafa Dinc
- Endocrinology and Metabolism, Kirklareli Training and Research Hospital, Kirklareli, TUR
| | - Deniz Oke
- Physical Medicine and Rehabilitation, Gaziosmanpasa Training and Research Hospital, Istanbul, TUR
| | | | - Burcak Karadeniz
- Rheumatology, Bagcilar Training and Research Hospital, Istanbul, TUR
| | - Gokhan Burul
- Internal Medicine, Bagcilar Training and Research Hospital, Istanbul, TUR
| | | |
Collapse
|
12
|
Yang X, Li X, Hu M, Huang J, Yu S, Zeng H, Mao L. EPA and DHA differentially improve insulin resistance by reducing adipose tissue inflammation-targeting GPR120/PPARγ pathway. J Nutr Biochem 2024; 130:109648. [PMID: 38631512 DOI: 10.1016/j.jnutbio.2024.109648] [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: 09/25/2023] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 04/19/2024]
Abstract
Insulin resistance (IR) is a global health challenge, often initiated by dysfunctional adipose tissue. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) may have different effects on IR, but the mechanisms are unknown. This study aims to evaluate the protective effect of EPA and DHA against IR in a high-fat diet (HFD) mice model and investigate whether EPA and DHA alter IR modulate the G-protein-poupled receptor 120/peroxisome proliferator-activated receptor γ (GPR120/PPARγ) pathway in macrophages and adipocytes, which may affect IR in adipocytes. The findings of this study show that 4% DHA had a better effect in improving IR and reducing inflammatory cytokines in adipose tissue of mice. Additionally, in the cell experiment, the use of AH7614 (a GPR120 antagonist) inhibited the glucose consumption increase and the increasable expression of PPARγ and insulin signaling molecules mediated by DHA in adipocytes. Furthermore, GW9662 (a PPARγ antagonist) hindered the upregulation of glucose consumption and insulin signaling molecule expression induced by EPA and DHA in adipocytes. DHA exhibited significant effects in reducing the number of migrated cells and inflammation. The compounds AH7614 and GW9662 hindered the suppressive effects of EPA and DHA on macrophage-induced IR in adipocytes. These findings suggest that DHA has a stronger potential in improving IR in adipocytes through the GPR120/PPARγ pathway in macrophages, when compared to EPA.
Collapse
Affiliation(s)
- Xian Yang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xudong Li
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Manjiang Hu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Jie Huang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Siyan Yu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Huanting Zeng
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Limei Mao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
13
|
Savulescu-Fiedler I, Mihalcea R, Dragosloveanu S, Scheau C, Baz RO, Caruntu A, Scheau AE, Caruntu C, Benea SN. The Interplay between Obesity and Inflammation. Life (Basel) 2024; 14:856. [PMID: 39063610 PMCID: PMC11277997 DOI: 10.3390/life14070856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is an important condition affecting the quality of life of numerous patients and increasing their associated risk for multiple diseases, including tumors and immune-mediated disorders. Inflammation appears to play a major role in the development of obesity and represents a central point for the activity of cellular and humoral components in the adipose tissue. Macrophages play a key role as the main cellular component of the adipose tissue regulating the chronic inflammation and modulating the secretion and differentiation of various pro- and anti-inflammatory cytokines. Inflammation also involves a series of signaling pathways that might represent the focus for new therapies and interventions. Weight loss is essential in decreasing cardiometabolic risks and the degree of associated inflammation; however, the latter can persist for long after the excess weight is lost, and can involve changes in macrophage phenotypes that can ensure the metabolic adjustment. A clear understanding of the pathophysiological processes in the adipose tissue and the interplay between obesity and chronic inflammation can lead to a better understanding of the development of comorbidities and may ensure future targets for the treatment of obesity.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Razvan Mihalcea
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Radu Octavian Baz
- Clinical Laboratory of Radiology and Medical Imaging, “Sf. Apostol Andrei” County Emergency Hospital, 900591 Constanta, Romania
- Department of Radiology and Medical Imaging, Faculty of Medicine, “Ovidius” University, 900527 Constanta, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- “Prof. Dr. Matei Balș” National Institute for Infectious Diseases, 021105 Bucharest, Romania
| |
Collapse
|
14
|
Chen X, Chen W, Xu H, Tian Y, Wang X, Chen X, Li J, Luo S, Hao L. Disulfiram Improves Fat Graft Retention by Modulating Macrophage Polarization With Inhibition of NLRP3 Inflammasome-Mediated Pyroptosis. Aesthet Surg J 2024; 44:NP501-NP518. [PMID: 38567442 PMCID: PMC11177556 DOI: 10.1093/asj/sjae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Macrophage-mediated inflammatory response in the early post-grafting period restricts fat graft retention. Pyroptosis is a novel type of programmed cell death that extensively participates in inflammatory pathologies. OBJECTIVES This study sought to determine whether macrophage pyroptosis was activated during the inflammatory phase after fat grafting and to investigate the efficacy of a pyroptosis inhibitor, disulfiram (DSF), in fat graft retention. METHODS We established a C57BL/6 mice fat grafting model and then analyzed macrophage pyroptosis. DSF (50 mg/kg, every other day) was intraperitoneally injected starting 1 hour before fat grafting and continued for 14 days. An in vitro co-culture system was established in which mouse RAW264.7 macrophages were co-cultured with apoptotic adipocytes to further validate the findings of the in vivo studies and to explore the underlying mechanisms. RESULTS Here we reported that macrophage pyroptosis was activated in both fat grafts and in vitro co-culture models. DSF was found to be a potent pyroptosis inhibitor, promoting M2 macrophage polarization. In addition, DSF was demonstrated to enhance vascularization and graft retention. CONCLUSIONS Our results suggested that pyroptosis plays a crucial role in the inflammatory cascade within fat grafts. DSF, being a clinically available drug, could be translated into a clinically effective drug for improving fat graft survival by inhibiting macrophage pyroptosis, therefore inducing M2 macrophage polarization and promoting neovascularization.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lijun Hao
- Corresponding Author: Dr Lijun Hao, No. 23 Youzheng Street, Nangang District, Harbin, Heilongjiang, 150000 P. R. China. E-mail:
| |
Collapse
|
15
|
Yu S, Hou C, Zhang X, Wei Z. Mesencephalic astrocyte-derived neurotrophic factor ameliorates inflammatory response in polycystic ovary syndrome via inhibiting TLR4-NF-κB-NLRP3 pathway. Biochem Biophys Res Commun 2024; 707:149782. [PMID: 38493745 DOI: 10.1016/j.bbrc.2024.149782] [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: 02/05/2024] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder in women of reproductive age, which often leads to female infertility. Chronic inflammation is a significant factor in the development of PCOS. Our study aimed to explore the impact of mesencephalic astrocyte-derived neurotrophic factor (MANF), a scientifically validated anti-inflammatory factor, on 99 diagnosed PCOS patients. We also investigated its effects on PCOS mice induced with dehydroepiandrosterone (DHEA) and KGN cells induced with dihydrotestosterone (DHT). Our findings revealed a decrease in serum MANF levels in PCOS patients, which were negatively associated with serum tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) levels. The administration of recombinant human MANF (rhMANF) in PCOS mice demonstrated a decrease in pro-inflammatory cytokines and monocytes/macrophages in both peripheral blood and ovarian tissues. Furthermore, the inclusion of rhMANF notably ameliorated DHEA-induced ovarian dysfunction and fibrosis by negatively regulating the toll-like receptor 4 (TLR4)-nuclear factor kappa B (NF-κB)-NLR family, pyrin domain containing protein 3 (NLRP3) pathway. Additionally, in vitro experiments showed that the up-regulation of MANF offset DHT-induced inhibition of viability and apoptosis in KGN cells. Collectively, this study highlights the anti-inflammatory properties of MANF in PCOS and suggests its potential as a therapeutic approach for the management of PCOS.
Collapse
Affiliation(s)
- Shujun Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Chao Hou
- School of Basic Medical Science, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xinru Zhang
- School of Basic Medical Science, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Zhaolian Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
16
|
Kloc M, Halasa M, Ghobrial RM. Macrophage niche imprinting as a determinant of macrophage identity and function. Cell Immunol 2024; 399-400:104825. [PMID: 38648700 DOI: 10.1016/j.cellimm.2024.104825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/22/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Macrophage niches are the anatomical locations within organs or tissues consisting of various cells, intercellular and extracellular matrix, transcription factors, and signaling molecules that interact to influence macrophage self-maintenance, phenotype, and behavior. The niche, besides physically supporting macrophages, imposes a tissue- and organ-specific identity on the residing and infiltrating monocytes and macrophages. In this review, we give examples of macrophage niches and the modes of communication between macrophages and surrounding cells. We also describe how macrophages, acting against their immune defensive nature, can create a hospitable niche for pathogens and cancer cells.
Collapse
Affiliation(s)
- Malgorzata Kloc
- Houston Methodist Research Institute, Transplant Immunology, Houston, TX, USA; Houston Methodist Hospital, Department of Surgery, Houston, TX, USA; University of Texas, MD Anderson Cancer Center, Department of Genetics, Houston, TX, USA.
| | - Marta Halasa
- Houston Methodist Research Institute, Transplant Immunology, Houston, TX, USA; Houston Methodist Hospital, Department of Surgery, Houston, TX, USA
| | - Rafik M Ghobrial
- Houston Methodist Research Institute, Transplant Immunology, Houston, TX, USA; Houston Methodist Hospital, Department of Surgery, Houston, TX, USA
| |
Collapse
|
17
|
Wang Y, Chen G, Xu M, Cui Y, He W, Zeng H, Zeng T, Cheng R, Li X. Caspase-1 Deficiency Modulates Adipogenesis through Atg7-Mediated Autophagy: An Inflammatory-Independent Mechanism. Biomolecules 2024; 14:501. [PMID: 38672517 PMCID: PMC11048440 DOI: 10.3390/biom14040501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Obesity stands as a significant risk factor for type 2 diabetes, hyperlipidemia, and cardiovascular diseases, intertwining increased inflammation and decreased adipogenesis with metabolic disorders. Studies have highlighted the correlation between Caspase-1 and inflammation in obesity, elucidating its essential role in the biological functions of adipose tissue. However, the impact of Caspase-1 on adipogenesis and the underlying mechanisms remain largely elusive. In our study, we observed a positive correlation between Caspase-1 expression and obesity and its association with adipogenesis. In vivo experiments revealed that, under normal diet conditions, Caspase-1 deficiency improved glucose homeostasis, stimulated subcutaneous adipose tissue expansion, and enhanced adipogenesis. Furthermore, our findings indicate that Caspase-1 deficiency promotes the expression of autophagy-related proteins and inhibits autophagy with 3-MA or CQ blocked Caspase-1 deficiency-induced adipogenesis in vitro. Notably, Caspase-1 deficiency promotes adipogenesis via Atg7-mediated autophagy activation. In addition, Caspase-1 deficiency resisted against high-fat diet-induced obesity and glucose intolerance. Our study proposes the downregulation of Caspase-1 as a promising strategy for mitigating obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rui Cheng
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xi Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
18
|
González-Sánchez GD, Granados-López AJ, López-Hernández Y, Robles MJG, López JA. miRNAs as Interconnectors between Obesity and Cancer. Noncoding RNA 2024; 10:24. [PMID: 38668382 PMCID: PMC11055034 DOI: 10.3390/ncrna10020024] [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: 12/18/2023] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 04/29/2024] Open
Abstract
Obesity and cancer are a concern of global interest. It is proven that obesity may trigger the development or progression of some types of cancer; however, the connection by non-coding RNAs has not been totally explored. In the present review, we discuss miRNAs and lncRNAs dysregulation involved in obesity and some cancers, shedding light on how these conditions may exacerbate one another through the dysregulation of ncRNAs. lncRNAs have been reported as regulating microRNAs. An in silico investigation of lncRNA and miRNA interplay is presented. Our investigation revealed 44 upregulated and 49 downregulated lncRNAs in obesity and cancer, respectively. miR-375, miR-494-3p, miR-1908, and miR-196 were found interacting with 1, 4, 4 and 4 lncRNAs, respectively, which are involved in PPARγ cell signaling regulation. Additionally, miR-130 was found to be downregulated in obesity and reported as modulating 5 lncRNAs controlling PPARγ cell signaling. Similarly, miR-128-3p and miR-143 were found to be downregulated in obesity and cancer, interacting with 5 and 4 lncRNAs, respectively, associated with MAPK cell signaling modulation. The delicate balance between miRNA and lncRNA expression emerges as a critical determinant in the development of obesity-associated cancers, presenting these molecules as promising biomarkers. However, additional and deeper studies are needed to reach solid conclusions about obesity and cancer connection by ncRNAs.
Collapse
Affiliation(s)
- Grecia Denisse González-Sánchez
- Doctorate in Biosciences, University Center of Los Altos, University of Guadalajara, Tepatitlán de Morelos C.P. 47620, Mexico;
| | - Angelica Judith Granados-López
- Laboratory of microRNAs and Cancer, Academic Unit of Biological Sciences, Autonomous University of Zacatecas “Francisco García Salinas”, Zacatecas C.P. 98066, Mexico;
| | - Yamilé López-Hernández
- Laboratory of Proteomics and Metabolomics, Cátedras-CONACYT, Academic Unit of Biological Sciences, Autonomous University of Zacatecas “Francisco García Salinas”, Zacatecas C.P. 98066, Mexico;
| | - Mayra Judith García Robles
- Biotechnology Department of the Polytechnic, University of Zacatecas, Fresnillo, Zacatecas C.P. 99059, Mexico
| | - Jesús Adrián López
- Laboratory of microRNAs and Cancer, Academic Unit of Biological Sciences, Autonomous University of Zacatecas “Francisco García Salinas”, Zacatecas C.P. 98066, Mexico;
| |
Collapse
|
19
|
Luo M, Zhao F, Cheng H, Su M, Wang Y. Macrophage polarization: an important role in inflammatory diseases. Front Immunol 2024; 15:1352946. [PMID: 38660308 PMCID: PMC11039887 DOI: 10.3389/fimmu.2024.1352946] [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: 12/09/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Macrophages are crucial cells in the human body's innate immunity and are engaged in a variety of non-inflammatory reactions. Macrophages can develop into two kinds when stimulated by distinct internal environments: pro-inflammatory M1-like macrophages and anti-inflammatory M2-type macrophages. During inflammation, the two kinds of macrophages are activated alternatively, and maintaining a reasonably steady ratio is critical for maintaining homeostasis in vivo. M1 macrophages can induce inflammation, but M2 macrophages suppress it. The imbalance between the two kinds of macrophages will have a significant impact on the illness process. As a result, there are an increasing number of research being conducted on relieving or curing illnesses by altering the amount of macrophages. This review summarizes the role of macrophage polarization in various inflammatory diseases, including autoimmune diseases (RA, EAE, MS, AIH, IBD, CD), allergic diseases (allergic rhinitis, allergic dermatitis, allergic asthma), atherosclerosis, obesity and type 2 diabetes, metabolic homeostasis, and the compounds or drugs that have been discovered or applied to the treatment of these diseases by targeting macrophage polarization.
Collapse
Affiliation(s)
| | | | | | | | - Yuanmin Wang
- The Third Affiliated Hospital of Zunyi Medical University, The First People’s Hospital of Zunyi, Zunyi, Guizhou, China
| |
Collapse
|
20
|
Zhang Y, Zhang B, Sun X. The molecular mechanism of macrophage-adipocyte crosstalk in maintaining energy homeostasis. Front Immunol 2024; 15:1378202. [PMID: 38650945 PMCID: PMC11033412 DOI: 10.3389/fimmu.2024.1378202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Interactions between macrophages and adipocytes in adipose tissue are critical for the regulation of energy metabolism and obesity. Macrophage polarization induced by cold or other stimulations can drive metabolic reprogramming of adipocytes, browning, and thermogenesis. Accordingly, investigating the roles of macrophages and adipocytes in the maintenance of energy homeostasis is critical for the development of novel therapeutic approaches specifically targeting macrophages in metabolic disorders such as obesity. Current review outlines macrophage polarization not only regulates the release of central nervous system and inflammatory factors, but controls mitochondrial function, and other factor that induce metabolic reprogramming of adipocytes and maintain energy homeostasis. We also emphasized on how the adipocytes conversely motivate the polarization of macrophage. Exploring the interactions between adipocytes and macrophages may provide new therapeutic strategies for the management of obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Yudie Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
21
|
Wu Y, Ma Y. CCL2-CCR2 signaling axis in obesity and metabolic diseases. J Cell Physiol 2024; 239:e31192. [PMID: 38284280 DOI: 10.1002/jcp.31192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/10/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024]
Abstract
Obesity and metabolic diseases, such as insulin resistance, type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular ailments, represent formidable global health challenges, bearing considerable implications for both morbidity and mortality rates. It has become increasingly evident that chronic, low-grade inflammation plays a pivotal role in the genesis and advancement of these conditions. The involvement of C-C chemokine ligand 2 (CCL2) and its corresponding receptor, C-C chemokine receptor 2 (CCR2), has been extensively documented in numerous inflammatory maladies. Recent evidence indicates that the CCL2/CCR2 pathway extends beyond immune cell recruitment and inflammation, exerting a notable influence on the genesis and progression of metabolic syndrome. The present review seeks to furnish a comprehensive exposition of the CCL2-CCR2 signaling axis within the context of obesity and metabolic disorders, elucidating its molecular mechanisms, functional roles, and therapeutic implications.
Collapse
Affiliation(s)
- Yue Wu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Yanchun Ma
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
22
|
Xiang G, Guo S, Xing N, Du Q, Qin J, Gao H, Zhang Y, Wang S. Mangiferin, a Potential Supplement to Improve Metabolic Syndrome: Current Status and Future Opportunities. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:355-386. [PMID: 38533569 DOI: 10.1142/s0192415x24500150] [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: 03/28/2024]
Abstract
Metabolic syndrome (MetS) represents a considerable clinical and public health burden worldwide. Mangiferin (MF), a flavonoid compound present in diverse species such as mango (Mangifera indica L.), papaya (Pseudocydonia sinensis (Thouin) C. K. Schneid.), zhimu (Anemarrhena asphodeloides Bunge), and honeybush tea (Cyclopia genistoides), boasts a broad array of pharmacological effects. It holds promising uses in nutritionally and functionally targeted foods, particularly concerning MetS treatment. It is therefore pivotal to systematically investigate MF's therapeutic mechanism for MetS and its applications in food and pharmaceutical sectors. This review, with the aid of a network pharmacology approach complemented by this experimental studies, unravels possible mechanisms underlying MF's MetS treatment. Network pharmacology results suggest that MF treats MetS effectively through promoting insulin secretion, targeting obesity and inflammation, alleviating insulin resistance (IR), and mainly operating via the phosphatidylinositol 3 kinase (PI3K)/Akt, nuclear factor kappa-B (NF-[Formula: see text]B), microtubule-associated protein kinase (MAPK), and oxidative stress signaling pathways while repairing damaged insulin signaling. These insights provide a comprehensive framework to understand MF's potential mechanisms in treating MetS. These, however, warrant further experimental validation. Moreover, molecular docking techniques confirmed the plausibility of the predicted outcomes. Hereafter, these findings might form the theoretical bedrock for prospective research into MF's therapeutic potential in MetS therapy.
Collapse
Affiliation(s)
- Gelin Xiang
- State Key Laboratory of Southwestern, Chinese Medicine Resources, School of Ethnic Medicine, Chengdu, P. R. China
| | - Sa Guo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Nan Xing
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Qinyun Du
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Jing Qin
- State Key Laboratory of Southwestern, Chinese Medicine Resources, School of Ethnic Medicine, Chengdu, P. R. China
| | - Huimin Gao
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan 620010, P. R. China
| | - Yi Zhang
- State Key Laboratory of Southwestern, Chinese Medicine Resources, School of Ethnic Medicine, Chengdu, P. R. China
| | - Shaohui Wang
- State Key Laboratory of Southwestern, Chinese Medicine Resources, School of Ethnic Medicine, Chengdu, P. R. China
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan 620010, P. R. China
| |
Collapse
|
23
|
Nakagawa S, Fukui-Miyazaki A, Yoshida T, Ishii Y, Murata E, Taniguchi K, Ishizu A, Kasahara M, Tomaru U. Decreased Proteasomal Function Exacerbates Endoplasmic Reticulum Stress-Induced Chronic Inflammation in Obese Adipose Tissue. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00076-2. [PMID: 38423355 DOI: 10.1016/j.ajpath.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/24/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
Low-grade chronic inflammation contributes to both aging and the pathogenesis of age-related diseases. White adipose tissue (WAT) in obese individuals exhibits chronic inflammation, which is associated with obesity-related disorders. Aging exacerbates obesity-related inflammation in WAT; however, the molecular mechanisms underlying chronic inflammation and its exacerbation by aging remain unclear. Age-related decline in activity of the proteasome, a multisubunit proteolytic complex, has been implicated in age-related diseases. This study employed a mouse model with decreased proteasomal function that exhibits age-related phenotypes to investigate the impact of adipocyte senescence on WAT inflammation. Transgenic mice expressing proteasomal subunit β5t with weak chymotrypsin-like activity experience reduced lifespan and develop age-related phenotypes. Mice fed with a high-fat diet and experiencing proteasomal dysfunction exhibited increased WAT inflammation, increased infiltration of proinflammatory M1-like macrophages, and increased proinflammatory adipocytokine-like monocyte chemoattractant protein-1, plasminogen activator inhibitor-1, and tumor necrosis factor-α, which are all associated with activation of endoplasmic reticulum (ER) stress-related pathways. Impaired proteasomal activity also activated ER stress-related molecules and induced expression of proinflammatory adipocytokines in adipocyte-like cells differentiated from 3T3-L1 cells. Collective evidence suggests that impaired proteasomal activity increases ER stress and that subsequent inflammatory pathways play pivotal roles in WAT inflammation. Because proteasomal function declines with age, age-related proteasome impairment may be involved in obesity-related inflammation among elderly individuals.
Collapse
Affiliation(s)
- Shimpei Nakagawa
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Aya Fukui-Miyazaki
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Takuma Yoshida
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasushi Ishii
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Eri Murata
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Department of Fundamental Nursing, School of Nursing, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Koji Taniguchi
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Ishizu
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Masanori Kasahara
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan.
| |
Collapse
|
24
|
Zhao S, Hu Y, Yang B, Zhang L, Xu M, Jiang K, Liu Z, Wu M, Huang Y, Li P, Liang SJ, Sun X, Hide G, Lun ZR, Wu Z, Shen J. The transplant rejection response involves neutrophil and macrophage adhesion-mediated trogocytosis and is regulated by NFATc3. Cell Death Dis 2024; 15:75. [PMID: 38242872 PMCID: PMC10798984 DOI: 10.1038/s41419-024-06457-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
The anti-foreign tissue (transplant rejection) response, mediated by the immune system, has been the biggest obstacle to successful organ transplantation. There are still many enigmas regarding this process and some aspects of the underlying mechanisms driving the immune response against foreign tissues remain poorly understood. Here, we found that a large number of neutrophils and macrophages were attached to the graft during skin transplantation. Furthermore, both types of cells could autonomously adhere to and damage neonatal rat cardiomyocyte mass (NRCM) in vitro. We have demonstrated that Complement C3 and the receptor CR3 participated in neutrophils/macrophages-mediated adhesion and damage this foreign tissue (NRCM or skin grafts). We have provided direct evidence that the damage to these tissues occurs by a process referred to as trogocytosis, a damage mode that has never previously been reported to directly destroy grafts. We further demonstrated that this process can be regulated by NFAT, in particular, NFATc3. This study not only enriches an understanding of host-donor interaction in transplant rejection, but also provides new avenues for exploring the development of novel immunosuppressive drugs which prevent rejection during transplant therapy.
Collapse
Affiliation(s)
- Siyu Zhao
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Yunyi Hu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Bicheng Yang
- The Andrology Department, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Lichao Zhang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Meiyining Xu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Kefeng Jiang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Zhun Liu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
| | - Mingrou Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Yun Huang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Peipei Li
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Si-Jia Liang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Sun Yat-sen University, 74 Zhongshan 2 Rd, Guangzhou, 510080, China
| | - Xi Sun
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Geoff Hide
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, M5 4WT, UK
| | - Zhao-Rong Lun
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, M5 4WT, UK
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Zhongdao Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Jia Shen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China.
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
25
|
Michetti F, Di Sante G, Clementi ME, Valeriani F, Mandarano M, Ria F, Di Liddo R, Rende M, Romano Spica V. The Multifaceted S100B Protein: A Role in Obesity and Diabetes? Int J Mol Sci 2024; 25:776. [PMID: 38255850 PMCID: PMC10815019 DOI: 10.3390/ijms25020776] [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: 11/17/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
The S100B protein is abundant in the nervous system, mainly in astrocytes, and is also present in other districts. Among these, the adipose tissue is a site of concentration for the protein. In the light of consistent research showing some associations between S100B and adipose tissue in the context of obesity, metabolic disorders, and diabetes, this review tunes the possible role of S100B in the pathogenic processes of these disorders, which are known to involve the adipose tissue. The reported data suggest a role for adipose S100B in obesity/diabetes processes, thus putatively re-proposing the role played by astrocytic S100B in neuroinflammatory/neurodegenerative processes.
Collapse
Affiliation(s)
- Fabrizio Michetti
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta” SCITEC-CNR, L.go F. Vito 1, 00168 Rome, Italy;
- Department of Neuroscience, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168 Rome, Italy
- Department of Medicine, LUM University, 70010 Casamassima, Italy
- Genes, Via Venti Settembre 118, 00187 Roma, Italy
| | - Gabriele Di Sante
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 06132 Perugia, Italy; (G.D.S.); (M.R.)
| | - Maria Elisabetta Clementi
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta” SCITEC-CNR, L.go F. Vito 1, 00168 Rome, Italy;
| | - Federica Valeriani
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (F.V.); (V.R.S.)
| | - Martina Mandarano
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, 06132 Perugia, Italy;
| | - Francesco Ria
- Department of Translational Medicine and Surgery, Section of General Pathology, Catholic University of the Sacred Heart, 00168 Rome, Italy;
| | - Rosa Di Liddo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy;
| | - Mario Rende
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 06132 Perugia, Italy; (G.D.S.); (M.R.)
| | - Vincenzo Romano Spica
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (F.V.); (V.R.S.)
| |
Collapse
|
26
|
Zhou D, Lu P, Mo X, Yang B, Chen T, Yao Y, Xiong T, Yue L, Yang X. Ferroptosis and metabolic syndrome and complications: association, mechanism, and translational applications. Front Endocrinol (Lausanne) 2024; 14:1248934. [PMID: 38260171 PMCID: PMC10800994 DOI: 10.3389/fendo.2023.1248934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Metabolic syndrome is a medical condition characterized by several metabolic disorders in the body. Long-term metabolic disorders raise the risk of cardiovascular disease (CVD) and type 2 diabetes mellitus (T2DM). Therefore, it is essential to actively explore the aetiology of metabolic syndrome (MetS) and its comorbidities to provide effective treatment options. Ferroptosis is a new form of cell death that is characterized by iron overload, lipid peroxide accumulation, and decreased glutathione peroxidase 4(GPX4) activity, and it involves the pathological processes of a variety of diseases. Lipid deposition caused by lipid diseases and iron overload is significant in metabolic syndrome, providing the theoretical conditions for developing ferroptosis. Recent studies have found that the major molecules of ferroptosis are linked to common metabolic syndrome consequences, such as T2DM and atherosclerosis. In this review, we first discussed the mechanics of ferroptosis, the regulatory function of inducers and inhibitors of ferroptosis, and the significance of iron loading in MetS. Next, we summarized the role of ferroptosis in the pathogenesis of MetS, such as obesity, type 2 diabetes, and atherosclerosis. Finally, we discussed relevant ferroptosis-targeted therapies and raised some crucial issues of concern to provide directions for future Mets-related treatments and research.
Collapse
Affiliation(s)
- Dongmei Zhou
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Peipei Lu
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xianglai Mo
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Bing Yang
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Ting Chen
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - You Yao
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Tian Xiong
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Lin Yue
- School of Nursing, Hunan University of Medicine, Huaihua, China
| | - Xi Yang
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| |
Collapse
|
27
|
Neagu M, Dobre EG. New Insights into the Link Between Melanoma and Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:851-867. [PMID: 39287874 DOI: 10.1007/978-3-031-63657-8_28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The significant increase in the incidence of obesity represents a global health crisis. Obesity is actually a multi-organ disease affecting the entire organism; hence, skin is no exception. As the functional alterations in the adipose tissue are contributing factors to many diseases, including cancer, recently, the link between the development of melanoma skin cancer and obesity gains increased attention. Besides several other factors, the increase of adipose stromal/stem cells (ASCs) impacts cancer progression. Moreover, increased production of cytokines and growth factors done by ASCs induces tumorigenesis and metastasis. The chronic inflammatory state that is sustained by this metabolic imbalance favors skin malignancies, melanoma included. Cutaneous melanoma, as an aggressive skin cancer, has both intrinsic and extrinsic risk factors where sun exposure and lifestyles are the main environmental factors inducing this skin cancer. With the advent of recent targeted and immune-based therapies in melanoma, the link between obesity and the efficacy of these therapies in melanoma remains controversial. A recent molecular relationship between the melanocortin pathway appending to both melanin synthesis and obesity was established. The biology of adipokines, molecules secreted by the adipose tissue, is linked to inflammation, and their molecular pathways can be involved in angiogenesis, migration, invasion, and proliferation of melanoma cells. In melanoma cells, among the most noticeable metabolic reprogramming characteristics is an increased rate of lipid synthesis. Lipid mediators impact classical oncogenic pathways, affecting melanoma progression. The chapter will tackle also the practical implications for melanoma prevention and treatment, namely, how metabolic manipulation can be exploited to overcome immunosuppression and support immune checkpoint blockade efficacy.
Collapse
Affiliation(s)
- Monica Neagu
- Immunology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
- Pathology Department, Colentina University Hospital, Bucharest, Romania
- Doctoral School of Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Elena-Georgiana Dobre
- Immunology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| |
Collapse
|
28
|
Kang Y, Ren P, Shen X, Kuang X, Yang X, Liu H, Yan H, Yang H, Kang X, Ding Z, Luo X, Ma J, Yang Y, Fan W. A Newly Synbiotic Combination Alleviates Obesity by Modulating the Gut Microbiota-Fat Axis and Inhibiting the Hepatic TLR4/NF-κB Signaling Pathway. Mol Nutr Food Res 2023; 67:e2300141. [PMID: 37594720 DOI: 10.1002/mnfr.202300141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/11/2023] [Indexed: 08/19/2023]
Abstract
SCOPE Obesity has been recognized as a worldwide public health crisis, this is accompanied by dysregulation of the intestinal microbiota and upregulation of liver steatosis and adipose inflammation. Synbiotic as a novel alternative therapy for obesity have recently gained much attention. METHODS This study innovatively research the anti-obesity properties of a newly synbiotic composed of Lactobacillus acidophilus, Bifidobacterium infantis and konjac glucomannan oligosaccharides. RESULTS The synbiotic treatment can reduce body weight, fat mass, blood sugar, liver steatosis and adipose inflammation in obesity mice fed by high-fat diet (HFD). Meanwhile, synbiotic treatment activated brown adipose tissue and improve energy, glucose and lipid metabolism. In addition, synbiotic treatment not solely enhanced the protection of intestinal barrier, but also ameliorated gut microbiota dysbiosis directly by enhancing beneficial microbes and reducing potentially harmful bacteria. Furthermore, the microbiome phenotype and functional prediction showed that synbiotic treatment can improve the gut microbiota functions involving inflammatory state, immune response, metabolism and pathopoiesia. CONCLUSION The synbiotic may be an effective candidate treatment strategy for the clinical prevention and treatment of obesity and other associated metabolic diseases such as hyperlipidemia, nonalcoholic fatty liver diseases by alleviating inflammatory response, regulating energy metabolism and maintaining the balance of intestinal microecology.
Collapse
Affiliation(s)
- Yongbo Kang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Peng Ren
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xiaorong Shen
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xiaoyu Kuang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xiaodan Yang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Haixia Liu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Huan Yan
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Hao Yang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xing Kang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Zeyuan Ding
- Laboratory of Morphology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xuguang Luo
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jieqiong Ma
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Ying Yang
- Department of Endocrinology, Affiliated Hospital of Yunnan University, Kunming, Yunnan, 650021, China
| | - Weiping Fan
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| |
Collapse
|
29
|
Lee J, Jeon S, Lee M, Yoon M. Fenofibrate alleviates insulin resistance by reducing tissue inflammation in obese ovariectomized mice. Nutr Diabetes 2023; 13:19. [PMID: 37935669 PMCID: PMC10630285 DOI: 10.1038/s41387-023-00249-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Fenofibrate is a hypolipidemic peroxisome proliferator-activated receptor α (PPARα) agonist used clinically to reduce hypercholesterolemia and hypertriglyceridemia. OBJECTIVE We investigated the effects of fenofibrate on insulin resistance and tissue inflammation in a high-fat diet (HFD)-fed ovariectomized (OVX) C57BL/6J mice, a mouse model of obese postmenopausal women. METHODS Female OVX mice were randomly divided into 3 groups and received a low-fat diet, an HFD, or an HFD supplemented with 0.05% (w/w) fenofibrate for 9 weeks. Parameters of insulin resistance and tissue inflammation were measured using blood analysis, histological analysis, immunohistochemistry, and quantitative real-time polymerase chain reaction. RESULTS When fenofibrate was administered to HFD-fed OVX mice for 9 weeks, we observed reductions in body weight gain, adipose tissue mass, and the size of visceral adipocytes without the change of food intake. Fenofibrate improved mild hyperglycemia, severe hyperinsulinemia, and glucose tolerance in these mice. It also reduced pancreatic islet size and insulin-positive β-cell area to levels similar to those in OVX mice fed a low-fat diet. Concomitantly, administration of fenofibrate not only suppressed pancreatic lipid accumulation but also decreased CD68-positive macrophages in both the pancreas and visceral adipose tissue. Treatment with fenofibrate reduced tumor necrosis factor α (TNFα) mRNA levels in adipose tissue and lowered serum TNFα levels. CONCLUSION These results suggest that fenofibrate treatment attenuates insulin resistance in part by reducing tissue inflammation and TNFα expression in HFD-fed OVX mice.
Collapse
Affiliation(s)
- Jungu Lee
- Department of Biological Sciences, Mokwon University, Daejeon, 35349, Korea
| | - Suyeon Jeon
- Department of Biological Sciences, Mokwon University, Daejeon, 35349, Korea
| | - Mijeong Lee
- Department of Biological Sciences, Mokwon University, Daejeon, 35349, Korea
| | - Michung Yoon
- Department of Biological Sciences, Mokwon University, Daejeon, 35349, Korea.
| |
Collapse
|
30
|
Sommer J, Ehnis H, Seitz T, Schneider J, Wild AB, Moceri S, Buechler C, Bozec A, Weber GF, Merkel S, Beckervordersandforth R, Steinkasserer A, Schüle R, Trebicka J, Hartmann A, Bosserhoff A, von Hörsten S, Dietrich P, Hellerbrand C. Four-and-a-Half LIM-Domain Protein 2 (FHL2) Induces Neuropeptide Y (NPY) in Macrophages in Visceral Adipose Tissue and Promotes Diet-Induced Obesity. Int J Mol Sci 2023; 24:14943. [PMID: 37834391 PMCID: PMC10573629 DOI: 10.3390/ijms241914943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Obesity is characterized by the expansion of the adipose tissue, usually accompanied by inflammation, with a prominent role of macrophages infiltrating the visceral adipose tissue (VAT). This chronic inflammation is a major driver of obesity-associated comorbidities. Four-and-a-half LIM-domain protein 2 (FHL2) is a multifunctional adaptor protein that is involved in the regulation of various biological functions and the maintenance of the homeostasis of different tissues. In this study, we aimed to gain new insights into the expression and functional role of FHL2 in VAT in diet-induced obesity. We found enhanced FHL2 expression in the VAT of mice with Western-type diet (WTD)-induced obesity and obese humans and identified macrophages as the cellular source of enhanced FHL2 expression in VAT. In mice with FHL2 deficiency (FHL2KO), WTD feeding resulted in reduced body weight gain paralleled by enhanced energy expenditure and uncoupling protein 1 (UCP1) expression, indicative of activated thermogenesis. In human VAT, FHL2 was inversely correlated with UCP1 expression. Furthermore, macrophage infiltration and the expression of the chemokine MCP-1, a known promotor of macrophage accumulation, was significantly reduced in WTD-fed FHL2KO mice compared with wild-type (wt) littermates. While FHL2 depletion did not affect the differentiation or lipid metabolism of adipocytes in vitro, FHL2 depletion in macrophages resulted in reduced expressions of MCP-1 and the neuropeptide Y (NPY). Furthermore, WTD-fed FHL2KO mice showed reduced NPY expression in VAT compared with wt littermates, and NPY expression was enhanced in VAT resident macrophages of obese individuals. Stimulation with recombinant NPY induced not only UCP1 expression and lipid accumulation but also MCP-1 expression in adipocytes. Collectively, these findings indicate that FHL2 is a positive regulator of NPY and MCP-1 expression in macrophages and herewith closely linked to the mechanism of obesity-associated lipid accumulation and inflammation in VAT. Thus, FHL2 appears as a potential novel target to interfere with the macrophage-adipocyte crosstalk in VAT for treating obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Judith Sommer
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstr. 17, D-91054 Erlangen, Germany; (J.S.); (H.E.); (T.S.); (J.S.); (R.B.); (A.B.); (P.D.)
| | - Hanna Ehnis
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstr. 17, D-91054 Erlangen, Germany; (J.S.); (H.E.); (T.S.); (J.S.); (R.B.); (A.B.); (P.D.)
| | - Tatjana Seitz
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstr. 17, D-91054 Erlangen, Germany; (J.S.); (H.E.); (T.S.); (J.S.); (R.B.); (A.B.); (P.D.)
| | - Julia Schneider
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstr. 17, D-91054 Erlangen, Germany; (J.S.); (H.E.); (T.S.); (J.S.); (R.B.); (A.B.); (P.D.)
| | - Andreas B. Wild
- Department of Immune Modulation, University Hospital Erlangen, Hartmannstr. 4, D-91052 Erlangen, Germany; (A.B.W.); (A.S.)
| | - Sandra Moceri
- Department of Experimental Therapy, Preclinical Experimental Center, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Palmsanlage 5, D-91054 Erlangen, Germany; (S.M.); (S.v.H.)
| | - Christa Buechler
- Department of Internal Medicine I, University Hospital of Regensburg, Franz-Josef-Strauß-Allee 11, D-93053 Regensburg, Germany;
| | - Aline Bozec
- Department of Internal Medicine 3, Rheumatology and Immunology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Glückstr. 6, D-91054 Erlangen, Germany;
| | - Georg F. Weber
- Department of Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Krankenhausstr. 12, D-91054 Erlangen, Germany; (G.F.W.)
| | - Susanne Merkel
- Department of Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Krankenhausstr. 12, D-91054 Erlangen, Germany; (G.F.W.)
| | - Ruth Beckervordersandforth
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstr. 17, D-91054 Erlangen, Germany; (J.S.); (H.E.); (T.S.); (J.S.); (R.B.); (A.B.); (P.D.)
| | - Alexander Steinkasserer
- Department of Immune Modulation, University Hospital Erlangen, Hartmannstr. 4, D-91052 Erlangen, Germany; (A.B.W.); (A.S.)
| | - Roland Schüle
- Center for Clinical Research, University of Freiburg Medical School, Breisacherstr. 66, D-79106 Freiburg, Germany;
| | - Jonel Trebicka
- Department of Internal Medicine B, University of Münster, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany;
| | - Arndt Hartmann
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, Krankenhausstr. 8/10, D-91054 Erlangen, Germany;
| | - Anja Bosserhoff
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstr. 17, D-91054 Erlangen, Germany; (J.S.); (H.E.); (T.S.); (J.S.); (R.B.); (A.B.); (P.D.)
| | - Stephan von Hörsten
- Department of Experimental Therapy, Preclinical Experimental Center, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Palmsanlage 5, D-91054 Erlangen, Germany; (S.M.); (S.v.H.)
| | - Peter Dietrich
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstr. 17, D-91054 Erlangen, Germany; (J.S.); (H.E.); (T.S.); (J.S.); (R.B.); (A.B.); (P.D.)
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstr. 17, D-91054 Erlangen, Germany; (J.S.); (H.E.); (T.S.); (J.S.); (R.B.); (A.B.); (P.D.)
| |
Collapse
|
31
|
Ruskovska T, Bernlohr DA. The Role of NAD + in Metabolic Regulation of Adipose Tissue: Implications for Obesity-Induced Insulin Resistance. Biomedicines 2023; 11:2560. [PMID: 37761000 PMCID: PMC10526756 DOI: 10.3390/biomedicines11092560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/31/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Obesity-induced insulin resistance is among the key factors in the development of type 2 diabetes, atherogenic dyslipidemia and cardiovascular disease. Adipose tissue plays a key role in the regulation of whole-body metabolism and insulin sensitivity. In obesity, adipose tissue becomes inflamed and dysfunctional, exhibiting a modified biochemical signature and adipokine secretion pattern that promotes insulin resistance in peripheral tissues. An important hallmark of dysfunctional obese adipose tissue is impaired NAD+/sirtuin signaling. In this chapter, we summarize the evidence for impairment of the NAD+/sirtuin pathway in obesity, not only in white adipose tissue but also in brown adipose tissue and during the process of beiging, together with correlative evidence from human studies. We also describe the role of PARPs and CD38 as important NAD+ consumers and discuss findings from experimental studies that investigated potential NAD+ boosting strategies and their efficacy in restoring impaired NAD+ metabolism in dysfunctional obese adipose tissue. In sum, these studies suggest a critical role of NAD+ metabolism in adipose biology and provide a basis for the potential development of strategies to restore metabolic health in obesity.
Collapse
Affiliation(s)
- Tatjana Ruskovska
- Faculty of Medical Sciences, Goce Delcev University, 2000 Stip, North Macedonia;
| | - David A. Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota—Twin Cities, Minneapolis, MN 55455, USA
| |
Collapse
|
32
|
Qiu L, Gao C, Wang H, Ren Y, Li J, Li M, Du X, Li W, Zhang J. Effects of dietary polyphenol curcumin supplementation on metabolic, inflammatory, and oxidative stress indices in patients with metabolic syndrome: a systematic review and meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne) 2023; 14:1216708. [PMID: 37522129 PMCID: PMC10376715 DOI: 10.3389/fendo.2023.1216708] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Objective The aim was to conduct a systematic review and meta-analysis for assessing the effectiveness and safety of dietary polyphenol curcumin supplement on metabolic, inflammatory, and oxidative stress indices in patients with metabolic syndrome (MetS). Methods A comprehensive search for clinical trials was conducted in the following scientific databases: PubMed, SCOPUS, Cochrane Library, EMBASE, Web of Science, and China Biological Medicine. Randomized controlled trials (RCTs) evaluating the efficacy and safety of curcumin supplement for MetS were identified. A random-effects meta-analysis was performed using inverse variance, and efficacy was expressed as mean difference (MD) with 95% confidence interval (CI). The metabolic syndrome markers that were evaluated in the present study included waist circumference (WC), fasting blood sugar (FBS), systolic blood pressure (SBP), diastolic blood pressure (DBP), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), tumor necrosis factor-a (TNF-a), interleukin 6 (IL-6), C-reactive protein (CRP), ultrasensitive c-reactive protein (hsCRP), and malondialdehyde (MDA). By employing the Cochrane tool, RCTs were assessed for bias risk. Results A total of 785 participants from 13 RCTs were included, with intervention durations ranging from 4 to 12 weeks. Compared with the control group, the curcumin group had positive effects on WC (MD = -2.16, 95% CI: -3.78 to -0.54, p = 0.009, seven studies), FBS (MD = -8.6, 95% CI: -15.45 to -1.75, p = 0.01, nine studies), DBP (MD = -2.8, 95% CI: -4.53 to - 1.06, p = 0.002, five studies), HDL-C (MD = 4.98, 95% CI: 2.58 to 7.38, p < 0.0001, eight studies), TNF-a (MD = -12.97, 95% CI: -18.37 to -7.57, p < 0.00001, two studies), CRP (MD = - 1.24, 95% CI: -1.71 to -0.77, p < 0.00001, two studies), and MDA (MD = -2.35, 95% CI: -4.47 to -0.24, p = 0.03, three studies). These improvements were statistically significant. Meanwhile, there was no significant improvement in SBP (MD = -4.82, 95% CI: -9.98 to 0.35, p = 0.07, six studies), TG (MD = 1.28, 95% CI: -3.75 to 6.30, p = 0.62, eight studies), IL-6 (MD = -1.5, 95% CI: -3.97 to 0.97, p = 0.23, two studies), or hsCRP (MD = -1.10, 95% CI: -4.35 to 2.16, p < 0.51, two studies). FBS, SBP, HDL-C, IL-6, CRP, hsCRP, and MDA had a relatively high heterogeneity. Conclusion Curcumin exhibited promising potential in enhancing markers associated with metabolic syndrome, including inflammation. However, additional studies are required to confirm such findings since the included evidence is limited and has a relatively high heterogeneity. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42022362553.
Collapse
Affiliation(s)
- Linjie Qiu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chunyang Gao
- Department of Special Needs International Medical, Peking University International Hospital, Beijing, China
| | - Haonan Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan Ren
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jixin Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Meijie Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinlei Du
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenjie Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jin Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
33
|
Paneru BD, Hill DA. The role of extracellular vesicle-derived miRNAs in adipose tissue function and metabolic health. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00027. [PMID: 37501663 PMCID: PMC10371064 DOI: 10.1097/in9.0000000000000027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023]
Abstract
Extracellular vesicles (EVs) are nanometer size lipid particles that are released from virtually every cell type. Recent studies have shown that miRNAs carried by EVs play important roles in intercellular and interorgan communication. In the context of obesity and insulin resistance, EV-derived miRNAs functionally bridge major metabolic organs, including the adipose tissue, skeletal muscle, liver, and pancreas, to regulate insulin secretion and signaling. As a result, many of these EV-derived miRNAs have been proposed as potential disease biomarkers and/or therapeutic agents. However, the field's knowledge of EV miRNA-mediated regulation of mammalian metabolism is still in its infancy. Here, we review the evidence indicating that EV-derived miRNAs provide cell-to-cell and organ-to-organ communication to support metabolic health, highlight the potential medical relevance of these discoveries, and discuss the most important knowledge gaps and future directions for this field.
Collapse
Affiliation(s)
- Bam D. Paneru
- Division of Allergy and Immunology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - David A. Hill
- Division of Allergy and Immunology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Institute for Immunology, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
34
|
Yang J, He J, Feng Y, Xiang M. Obesity contributes to hepatocellular carcinoma development via immunosuppressive microenvironment remodeling. Front Immunol 2023; 14:1166440. [PMID: 37266440 PMCID: PMC10231659 DOI: 10.3389/fimmu.2023.1166440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/05/2023] [Indexed: 06/03/2023] Open
Abstract
It is generally recognized that the initiation of obesity-related hepatocellular carcinoma (HCC) is closely associated with hepatic inflammation. However, the paradoxical role of inflammation in the initiation and progression of HCC is highlighted by the fact that the inflammatory HCC is accompanied by significant immune effector cells infiltration compared to non-inflammatory HCC and HCC with enhanced immune response exhibits better survival. Importantly, the cancer progression has been primarily attributed to the immunosuppression, which can also be induced by obesity. Furthermore, the increased risk of viral infection and thus viral-HCC in obese individuals supports the view that obesity contributes to HCC via immunosuppression. Here, we have reviewed the various mechanisms responsible for obesity-induced tumor immune microenvironment and immunosuppression in obesity-related HCC. We highlight that the obesity-induced immunosuppression originates from lipid disorder as well as metabolic reprogramming and propose potential therapeutic strategy for HCC based on the current success of immunotherapy.
Collapse
|
35
|
Bauer S, Hezinger L, Rexhepi F, Ramanathan S, Kufer TA. NOD-like Receptors-Emerging Links to Obesity and Associated Morbidities. Int J Mol Sci 2023; 24:ijms24108595. [PMID: 37239938 DOI: 10.3390/ijms24108595] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity and its associated metabolic morbidities have been and still are on the rise, posing a major challenge to health care systems worldwide. It has become evident over the last decades that a low-grade inflammatory response, primarily proceeding from the adipose tissue (AT), essentially contributes to adiposity-associated comorbidities, most prominently insulin resistance (IR), atherosclerosis and liver diseases. In mouse models, the release of pro-inflammatory cytokines such as TNF-alpha (TNF-α) and interleukin (IL)-1β and the imprinting of immune cells to a pro-inflammatory phenotype in AT play an important role. However, the underlying genetic and molecular determinants are not yet understood in detail. Recent evidence demonstrates that nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family proteins, a group of cytosolic pattern recognition receptors (PRR), contribute to the development and control of obesity and obesity-associated inflammatory responses. In this article, we review the current state of research on the role of NLR proteins in obesity and discuss the possible mechanisms leading to and the outcomes of NLR activation in the obesity-associated morbidities IR, type 2 diabetes mellitus (T2DM), atherosclerosis and non-alcoholic fatty liver disease (NAFLD) and discuss emerging ideas about possibilities for NLR-based therapeutic interventions of metabolic diseases.
Collapse
Affiliation(s)
- Sarah Bauer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| | - Lucy Hezinger
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| | - Fjolla Rexhepi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Thomas A Kufer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| |
Collapse
|
36
|
Zhang Z, Wang A, Wang Y, Sun W, Zhou X, Xu Q, Mao L, Zhang J. Canthin-6-Ones: Potential Drugs for Chronic Inflammatory Diseases by Targeting Multiple Inflammatory Mediators. Molecules 2023; 28:3381. [PMID: 37110614 PMCID: PMC10141368 DOI: 10.3390/molecules28083381] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Chronic inflammatory disease (CID) is a category of medical conditions that causes recurrent inflammatory attacks in multiple tissues. The occurrence of CID is related to inappropriate immune responses to normal tissue substances and invading microbes due to many factors, such as defects in the immune system and imbalanced regulation of commensal microbes. Thus, effectively keeping the immune-associated cells and their products in check and inhibiting aberrant activation of the immune system is a key strategy for the management of CID. Canthin-6-ones are a subclass of β-carboline alkaloids isolated from a wide range of species. Several emerging studies based on in vitro and in vivo experiments reveal that canthin-6-ones may have potential therapeutic effects on many inflammatory diseases. However, no study has yet summarized the anti-inflammatory functions and the underlying mechanisms of this class of compounds. This review provides an overview of these studies, focusing on the disease entities and the inflammatory mediators that have been shown to be affected by canthin-6-ones. In particular, the major signaling pathways affected by canthin-6-ones, such as the NLR family pyrin domain containing 3 (NLRP3) inflammasome and the NF-κB signaling pathway, and their roles in several CIDs are discussed. Moreover, we discuss the limitations in studies of canthin-6-ones and provide possible solutions. In addition, a perspective that may suggest possible future research directions is provided. This work may be helpful for further mechanistic studies and possible therapeutic applications of canthin-6-ones in the treatment of CID.
Collapse
Affiliation(s)
- Zongying Zhang
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Anqi Wang
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Yunhan Wang
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Weichen Sun
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Qiuyun Xu
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Liming Mao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong 226019, China
| | - Jie Zhang
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| |
Collapse
|
37
|
Hancková M, Miháliková L, Pastoreková S, Betáková T. Hypoxia alters the immune response in mouse peritoneal macrophages infected with influenza a virus with truncated NS1 protein. Cytokine 2023; 164:156138. [PMID: 36796258 DOI: 10.1016/j.cyto.2023.156138] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/30/2022] [Accepted: 01/19/2023] [Indexed: 02/16/2023]
Abstract
Macrophages are the most abundant cells in infected tissue and are involved in the clearing infection, and immunomodulation of the innate and adaptive immune response. NS80 virus of influenza A virus, which encodes only the first 80 aa of the NS1 protein, suppresses the immune host response and is associated with enhanced pathogenicity. Hypoxia promotes infiltration of peritoneal macrophages into the adipose tissue and production of cytokines. To understand the role of hypoxia in the regulation of immune response, macrophages were infected with A/WSN/33 (WSN) and NS80 virus, and transcriptional profiles of the RIG-I-like receptor signalling pathway and expression of cytokines were evaluated in normoxia and hypoxia. Hypoxia inhibited the proliferation of IC-21 cells, downregulated the RIG-I-like receptor signalling pathway, and inhibited transcriptional activity of IFN-α, IFN-β, IFN-ε, and IFN-λ mRNA in infected macrophages. While transcription of IL-1β and Casp-1 mRNAs were increased in infected macrophages in normoxia, hypoxia resulted in decreased transcription activity of IL-1β and Casp-1 mRNAs. Hypoxia significantly affected expression of the translation factors IRF4, IFN-γ, and CXCL10 involved in regulation of immune response and polarization of the macrophages. The expression of pro-inflammatory cytokines such as sICAM-1, IL-1α, TNF-α, CCL2, CCL3, CXCL12, and M-CSF was to a large extent affected in uninfected and infected macrophages cultivated in hypoxia. The NS80 virus increased the expression of M-CSF, IL-16, CCL2, CCL3, and CXCL12, especially under hypoxia. The results show that hypoxia may play an important role in peritoneal macrophage activation, regulates the innate and adaptive immune response, changes production of pro-inflammatory cytokines, promotes macrophage polarization, and could affect the function of other immune cells.
Collapse
Affiliation(s)
- Miriam Hancková
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Virology, Bratislava, Slovak Republic
| | - Lucia Miháliková
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Virology, Bratislava, Slovak Republic
| | - Silvia Pastoreková
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Virology, Bratislava, Slovak Republic
| | - Tatiana Betáková
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Virology, Bratislava, Slovak Republic; Comenius University in Bratislava, Faculty of Natural Sciences, Department of Microbiology and Virology, Bratislava, Slovak Republic.
| |
Collapse
|
38
|
Adipose tissue macrophages and their role in obesity-associated insulin resistance: an overview of the complex dynamics at play. Biosci Rep 2023; 43:232519. [PMID: 36718668 PMCID: PMC10011338 DOI: 10.1042/bsr20220200] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
Obesity, a major global health concern, is characterized by serious imbalance between energy intake and expenditure leading to excess accumulation of fat in adipose tissue (AT). A state of chronic low-grade AT inflammation is prevalent during obesity. The adipose tissue macrophages (ATM) with astounding heterogeneity and complex regulation play a decisive role in mediating obesity-induced insulin resistance. Adipose-derived macrophages were broadly classified as proinflammatory M1 and anti-inflammatory M2 subtypes but recent reports have proclaimed several novel and intermediate profiles, which are crucial in understanding the dynamics of macrophage phenotypes during development of obesity. Lipid-laden hypertrophic adipocytes release various chemotactic signals that aggravate macrophage infiltration into AT skewing toward mostly proinflammatory status. The ratio of M1-like to M2-like macrophages is increased substantially resulting in copious secretion of proinflammatory mediators such as TNFα, IL-6, IL-1β, MCP-1, fetuin-A (FetA), etc. further worsening insulin resistance. Several AT-derived factors could influence ATM content and activation. Apart from being detrimental, ATM exerts beneficial effects during obesity. Recent studies have highlighted the prime role of AT-resident macrophage subpopulations in not only effective clearance of excess fat and dying adipocytes but also in controlling vascular integrity, adipocyte secretions, and fibrosis within obese AT. The role of ATM subpopulations as friend or foe is determined by an intricate interplay of such factors arising within hyperlipidemic microenvironment of obese AT. The present review article highlights some of the key research advances in ATM function and regulation, and appreciates the complex dynamics of ATM in the pathophysiologic scenario of obesity-associated insulin resistance.
Collapse
|
39
|
Santillana N, Astudillo-Guerrero C, D’Espessailles A, Cruz G. White Adipose Tissue Dysfunction: Pathophysiology and Emergent Measurements. Nutrients 2023; 15:nu15071722. [PMID: 37049561 PMCID: PMC10096946 DOI: 10.3390/nu15071722] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
White adipose tissue (AT) dysfunction plays an important role in the development of cardiometabolic alterations associated with obesity. AT dysfunction is characterized by the loss of the expansion capacity of the AT, an increment in adipocyte hypertrophy, and changes in the secretion profile of adipose cells, associated with accumulation of macrophages and inflammation. Since not all people with an excess of adiposity develop comorbidities, it is necessary to find simple tools that can evidence AT dysfunction and allow the detection of those people with the potential to develop metabolic alterations. This review focuses on the current pathophysiological mechanisms of white AT dysfunction and emerging measurements to assess its functionality.
Collapse
Affiliation(s)
- Natalia Santillana
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago 8380453, Chile
| | - Camila Astudillo-Guerrero
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Amanda D’Espessailles
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| |
Collapse
|
40
|
Liu ZJ, Zhu CF. Causal relationship between insulin resistance and sarcopenia. Diabetol Metab Syndr 2023; 15:46. [PMID: 36918975 PMCID: PMC10015682 DOI: 10.1186/s13098-023-01022-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/08/2023] [Indexed: 03/15/2023] Open
Abstract
Sarcopenia is a multifactorial disease characterized by reduced muscle mass and function, leading to disability, death, and other diseases. Recently, the prevalence of sarcopenia increased considerably, posing a serious threat to health worldwide. However, no clear international consensus has been reached regarding the etiology of sarcopenia. Several studies have shown that insulin resistance may be an important mechanism in the pathogenesis of induced muscle attenuation and that, conversely, sarcopenia can lead to insulin resistance. However, the causal relationship between the two is not clear. In this paper, the pathogenesis of sarcopenia is analyzed, the possible intrinsic causal relationship between sarcopenia and insulin resistance examined, and research progress expounded to provide a basis for the clinical diagnosis, treatment, and study of the mechanism of sarcopenia.
Collapse
Affiliation(s)
- Zi-jian Liu
- Shenzhen Clinical Medical College, Southern Medical University, Guangdong, 518101 China
| | - Cui-feng Zhu
- Shenzhen Hospital of Southern Medical University, Guangdong, 518101 China
| |
Collapse
|
41
|
Xiao L, Mochizuki M, Wang D, Shimamura N, Sunada K, Nakahara T. Types of cell culture inserts affect cell crosstalk between co-cultured macrophages and adipocytes. Biochem Biophys Res Commun 2023; 658:10-17. [PMID: 37011478 DOI: 10.1016/j.bbrc.2023.03.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023]
Abstract
Cell culture inserts offer an in vivo-like microenvironment to investigate cell-cell interactions between co-cultivated cells. However, it is unclear if types of inserts affect cell crosstalk. Here, we developed an environment-friendly cell culture insert, XL-insert, which can reduce plastic waste with lower cost. We compared XL insert with two types of commercial disposable culture inserts, Koken® insert with atelocollagen membrane (Col-inserts) and Falcon® inserts with plastic membrane (PET-inserts) on cell-cell interactions in co-cultivated THP-1 macrophages and OP9 adipocytes. Scanning electron microscope, immunoassay and imaging analysis showed that among three types of inserts, XL-inserts allowed cytokines from co-cultivated macrophages and adipocytes to diffuse freely and offered preferable in vivo-like microenvironment for cell-cell interactions. PET-inserts showed limitations for intercellular communication due to some pores being blocked by somas on the membrane that caused much lower permeability for cytokines passing through. Col-inserts blocked large sized cytokines but allowed small sized molecules to permeate resulting in improved lipid accumulation and adiponectin secretion in OP9 adipocytes. Taken together, our data demonstrated that membrane type and pore size on the membrane affect the cross-talk between co-cultivated cells very differently. Some previous co-culture studies might have different results if the inserts were changed.
Collapse
Affiliation(s)
- Li Xiao
- Department of Pharmacology, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan.
| | - Mai Mochizuki
- Department of Life Science Dentistry, The Nippon Dental University, Tokyo, Japan; Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan.
| | - Dongliang Wang
- Beijing Xiaoxiandun Biotechnology Co., Ltd., No. 150, Guanzhuang Road, Changying Town, Chaoyang District, Beijing, 100020, China; Hebei Edible Bird's Nest Fresh Stew Technology Innovation Center, Bazhou Economic Development Zone, Langfang, 065700, China.
| | - Naohiro Shimamura
- Department of Dental Anesthesiology, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan.
| | - Katsuhisa Sunada
- Department of Dental Anesthesiology, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan.
| | - Taka Nakahara
- Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan.
| |
Collapse
|
42
|
7,8-Dihydroxyflavone Attenuates Inflammatory Response and Insulin Resistance Induced by the Paracrine Interaction between Adipocytes and Macrophages. Int J Mol Sci 2023; 24:ijms24043520. [PMID: 36834930 PMCID: PMC9961847 DOI: 10.3390/ijms24043520] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Obesity-induced inflammation and insulin resistance are mediated by macrophage infiltration into adipose tissue. We investigated the effects of 7,8-dihydroxyflavone (7,8-DHF), a flavone found in plants, on the inflammatory response and insulin resistance induced by the interaction between adipocytes and macrophages. Hypertrophied 3T3-L1 adipocytes were cocultured with RAW 264.7 macrophages and treated with 7,8-DHF (3.12, 12.5, and 50 μM). The inflammatory cytokines and free fatty acid (FFA) release were evaluated by assay kits, and signaling pathways were determined by immunoblotting. Coculture of adipocytes and macrophages increased inflammatory mediators, such as nitric oxide (NO), monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6) and FFA secretion but suppressed the production of anti-inflammatory adiponectin. 7,8-DHF counteracted the coculture-induced changes (p < 0.001). 7,8-DHF also inhibited c-Jun N-terminal kinase (JNK) activation and blocked nuclear factor kappa B (NF-κB) nuclear translocation in the coculture system (p < 0.01). In addition, adipocytes cocultured with macrophages did not increase glucose uptake and Akt phosphorylation in response to insulin. However, 7,8-DHF treatment recovered the impaired responsiveness to insulin (p < 0.01). These findings show that 7,8-DHF alleviates inflammation and adipocyte dysfunction in the coculture of hypertrophied 3T3-L1 adipocytes and RAW 264.7 macrophages, indicating its potential as a therapeutic agent for obesity-induced insulin resistance.
Collapse
|
43
|
Rudyk M, Hurmach Y, Serhiichuk T, Akulenko I, Skivka L, Berehova T, Ostapchenko L. Multi-probiotic consumption sex-dependently interferes with MSG-induced obesity and concomitant phagocyte pro-inflammatory polarization in rats: Food for thought about personalized nutrition. Heliyon 2023; 9:e13381. [PMID: 36816299 PMCID: PMC9932736 DOI: 10.1016/j.heliyon.2023.e13381] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Epidemic scope which obesity has reached in many countries necessitates shifting the emphasis in medicine from traditional reaction to individualized and personalized prevention. Numerous trials convincingly prove sexual dimorphism of obesity in morbidity, pathophysiology, comorbidity, outcomes and prophylaxis efficacy. Obesity is characterized by chronic systemic low-grade inflammation that creates the preconditions for the emergence of numerous comorbidities. Leading role in the initiation, propagation and resolution of inflammation belongs to tissue resident and circulating phagocytes. The outcome of inflammation largely depends on phagocyte functional polarization, which in turn is governed by environmental stimuli. Gut microbiota (GM), whose disturbances are one of the key pathogenetic features in obesity, substantially affect phagocyte functions and can either aggravate or calm obesity-associated inflammation. Probiotics possess promising physiological functions, including microbiota-restoring and anti-inflammatory traits, that may possibly help prevent obesity. However, sex-specific effects of probiotic supplementation for targeted obesity prevention remain unknown. The aim of the current study was aimed to compare the effect of multi-probiotic preparation used in prophylactic regimen on the adiposity, profile of culturable GM and its short-chain fatty acids as well as on functional profile of phagocytes from different locations in male and female rats with monosodium glutamate (MSG)-induced obesity. Obesity was induced by neonatal MSG injections in male and female rats, who were given the multi-species probiotic during juvenile and adult developmental stages. Culturable fecal and mucosa-associated microbiota of the intestine were examined using selective diagnostic media. Short-chain fatty acid profile in fecal samples was determined by GC-MS. Phagocyte functional profile was evaluated using flow cytometry and colorimetric methods. Probiotic supplementation after the administration of MSG prevented weight gain and fat accumulation, inflammatory phagocyte activation and alterations in GM in female rats. In male MSG-injected rats, probiotic supplementation restricted but did not prevent weight gain and fat deposition, alleviated but did not prevent systemic inflammation, prevented the alterations in GM, but with residual imbalance in the ratio of obligate anaerobic to facultative anaerobic bacteria. Our findings emphasize the necessity of sex-centered approaches to the prophylactic use of probiotics in obesity in the context of predictive preventive and personalized medicine.
Collapse
Affiliation(s)
- Mariia Rudyk
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine,Corresponding author.
| | - Yevheniia Hurmach
- Bogomolets National Medical University, 13, T. Shevchenko Blvd, Kyiv, 01601, Ukraine
| | - Tetiana Serhiichuk
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Iryna Akulenko
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Larysa Skivka
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Tetiana Berehova
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Liudmyla Ostapchenko
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| |
Collapse
|
44
|
Yan J, Wang Y, Mu Z, Han X, Bi L, Wang X, Song P, Kang Y, Wang L, Zhang X, Wang Y, Zhang H. Gold Nanobipyramid-Mediated Apoptotic Camouflage of Adipocytes for Obesity Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207686. [PMID: 36502507 DOI: 10.1002/adma.202207686] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/22/2022] [Indexed: 06/17/2023]
Abstract
Obesity treatment is a global public health challenge due to inadequate weight loss and weight regain even after endeavors with multimodal treatments. Considering the abundance of resident macrophages in adipose tissues, precise regulation of the interactions between macrophages and adipocytes may provide chances for immunotherapy of obesity. Herein, inspired by the phagocytosis of macrophages to clear apoptotic cells in homeostasis, an immunotherapy strategy for obesity treatment is proposed for the first time through apoptotic camouflage of adipocytes by PA Au BPs to activate macrophages for clearance, where PA Au BPs are gold nanobipyramids engineered with adipose-targeting and apoptotic cell-mimicking functions. During clearance, the macrophage is switched from pro-inflammatory M1 to anti-inflammatory M2, remarkably modulating the immune microenvironment of adipose tissues to prevent weight regain. After inguinal injection with PA Au BPs, the body weights of obese mice are effectively decreased by 24.4% and can be decreased by 33.3% when combined with photothermal lipolysis, and little weight regain is associated with these treatments. This study demonstrates that the strategy of camouflaging adipocytes with apoptotic features holds great potential for obesity immunotherapy.
Collapse
Affiliation(s)
- Jiao Yan
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Yanjing Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Zhengzhi Mu
- Key Laboratory of Bionic Engineering, Ministry of Education, Jilin University, Changchun, 130022, China
| | - Xiaoqing Han
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Luopeng Bi
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin, 130022, China
| | - Xingbo Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Panpan Song
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yaqing Kang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Lulu Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Xueyan Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yanbo Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin, 130022, China
| | - Haiyuan Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
45
|
Sun K, Li X, Scherer PE. Extracellular Matrix (ECM) and Fibrosis in Adipose Tissue: Overview and Perspectives. Compr Physiol 2023; 13:4387-4407. [PMID: 36715281 PMCID: PMC9957663 DOI: 10.1002/cphy.c220020] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fibrosis in adipose tissue is a major driver of obesity-related metabolic dysregulation. It is characterized by an overaccumulation of extracellular matrix (ECM) during unhealthy expansion of adipose tissue in response to over nutrition. In obese adipose-depots, hypoxia stimulates multiple pro-fibrotic signaling pathways in different cell populations, thereby inducing the overproduction of the ECM components, including collagens, noncollagenous proteins, and additional enzymatic components of ECM synthesis. As a consequence, local fibrosis develops. The result of fibrosis-induced mechanical stress not only triggers cell necrosis and inflammation locally in adipose tissue but also leads to system-wide lipotoxicity and insulin resistance. A better understanding of the mechanisms underlying the obesity-induced fibrosis will help design therapeutic approaches to reduce or reverse the pathological changes associated with obese adipose tissue. Here, we aim to summarize the major advances in the field, which include newly identified fibrotic factors, cell populations that contribute to the fibrosis in adipose tissue, as well as novel mechanisms underlying the development of fibrosis. We further discuss the potential therapeutic strategies to target fibrosis in adipose tissue for the treatment of obesity-linked metabolic diseases and cancer. © 2023 American Physiological Society. Compr Physiol 13:4387-4407, 2023.
Collapse
Affiliation(s)
- Kai Sun
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xin Li
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Philipp E. Scherer
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
46
|
Asprosin Exerts Pro-Inflammatory Effects in THP-1 Macrophages Mediated via the Toll-like Receptor 4 (TLR4) Pathway. Int J Mol Sci 2022; 24:ijms24010227. [PMID: 36613673 PMCID: PMC9820073 DOI: 10.3390/ijms24010227] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Adipose tissue is a dynamic endocrine organ, secreting a plethora of adipokines which play a key role in regulating metabolic homeostasis and other physiological processes. An altered adipokine secretion profile from adipose tissue depots has been associated with obesity and related cardio-metabolic diseases. Asprosin is a recently described adipokine that is released in response to fasting and can elicit orexigenic and glucogenic effects. Circulating asprosin levels are elevated in a number of cardio-metabolic diseases, including obesity and type 2 diabetes. In vitro studies have reported pro-inflammatory effects of asprosin in a variety of tissues. The present study aimed to further elucidate the role of asprosin in inflammation by exploring its potential effect(s) in THP-1 macrophages. THP-1 monocytes were differentiated to macrophages by 48 h treatment with dihydroxyvitamin D3. Macrophages were treated with 100 nM recombinant human asprosin, 100 ng/mL lipopolysaccharide (LPS), and 10 μM caffeic acid phenethyl ester (CAPE; an inhibitor of NFκB activation) or 1 µM TAK-242 (a Toll-like receptor 4, TLR4, inhibitor). The expression and secretion of pertinent pro-inflammatory mediators were measured by qPCR, Western blot, ELISA and Bioplex. Asprosin stimulation significantly upregulated the expression and secretion of the pro-inflammatory cytokines: tumour necrosis factor α (TNFα), interleukin-1β (IL-1β), IL-8 and IL-12 in vitro. This pro-inflammatory response in THP-1 macrophages was partly attenuated by the treatments with CAPE and was significantly inhibited by TAK-242 treatment. Asprosin-induced inflammation is significantly counteracted by TLR4 inhibition in THP-1 macrophages, suggesting that asprosin exerts its pro-inflammatory effects, at least in part, via the TLR4 signalling pathway.
Collapse
|
47
|
Králová A, Kubátová H, Kauerová S, Janoušek L, Froněk J, Králová Lesná I, Poledne R. Cholesterol efflux and macrophage polarization in human adipose tissue. Physiol Res 2022. [DOI: 10.33549/physiolres.934926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The pro-inflammatory status of adipose tissue (AT) has been found to be related to reverse cholesterol transport (RCT) from peritoneal macrophages. However, this finding was made in experimental models using induced peritonitis and isolated peritoneal macrophages of animals. This experimental relationship is in agreement with RCT changes in man in two extreme situations, sepsis or cardiovascular complications.
Given the above, we sought to test RTC in relationship to macrophage polarization in the visceral AT (VAT) of living kidney donors (LKDs) and the effect of conditioned media obtained from their AT. The influence of ATCM on CE capacity was first assessed in an experiment where standard plasma was used as cholesterol acceptor from [14C] cholesterol labeled THP-1 cells. Conditioned media as a product of LKDs’ incubated AT showed no effect on CE. Likewise, we did not find any effect of individual plasma of LKDs on CE when individual plasma of LKDs were used as acceptors. On the other hand, we documented an effect of LKDs’ adipose cell size on CE. Our results indicate that the pro-inflammatory status of human AT is not likely induced by disrupted RCT but might be influenced by the metabolic status of LKDs’ adipose tissue.
Collapse
Affiliation(s)
| | | | | | | | | | | | - R Poledne
- Laboratory for Atherosclerosis Research, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| |
Collapse
|
48
|
Tremblay EJ, Tchernof A, Pelletier M, Chabot N, Joanisse DR, Mauriège P. Contribution of markers of adiposopathy and adipose cell size in predicting insulin resistance in women of varying age and adiposity. Adipocyte 2022; 11:175-189. [PMID: 35436409 PMCID: PMC9037496 DOI: 10.1080/21623945.2022.2059902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/07/2022] [Accepted: 03/27/2022] [Indexed: 12/30/2022] Open
Abstract
Adipose tissue (AT) dysfunctions, such as adipocyte hypertrophy, macrophage infiltration and secretory adiposopathy (low plasma adiponectin/leptin, A/L, ratio), associate with metabolic disorders. However, no study has compared the relative contribution of these markers to cardiometabolic risk in women of varying age and adiposity. Body composition, regional AT distribution, lipid-lipoprotein profile, glucose homeostasis and plasma A and L levels were determined in 67 women (age: 40-62 years; BMI: 17-41 kg/m2). Expression of macrophage infiltration marker CD68 and adipocyte size were measured from subcutaneous abdominal (SCABD) and omental (OME) fat. AT dysfunction markers correlated with most lipid-lipoprotein levels. The A/L ratio was negatively associated with fasting insulinemia and HOMA-IR, while SCABD or OME adipocyte size and SCABD CD68 expression were positively related to these variables. Combination of tertiles of largest adipocyte size and lowest A/L ratio showed the highest HOMA-IR. Multiple regression analyses including these markers and TAG levels revealed that the A/L ratio was the only predictor of fasting insulinemia and HOMA-IR. The contribution of the A/L ratio was superseded by adipose cell size in the model where the latter replaced TAGs. Finally, leptinemia was a better predictor of IR than adipocyte size and the A/L ratio in our participants sample.
Collapse
Affiliation(s)
- Eve-Julie Tremblay
- Département de kinésiologie, Faculté de médecine, Université Laval, Québec, Canada
- Centre de recherche de l’institut Universitaire de cardiologie et pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Canada
| | - André Tchernof
- Centre de recherche de l’institut Universitaire de cardiologie et pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Canada
- École de Nutrition, Faculté des sciences de l’agriculture et de l’alimentation, Université Laval, Québec, Canada
| | - Mélissa Pelletier
- Centre de recherche de l’institut Universitaire de cardiologie et pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Canada
| | - Nicolas Chabot
- Département de kinésiologie, Faculté de médecine, Université Laval, Québec, Canada
- Centre de recherche de l’institut Universitaire de cardiologie et pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Canada
| | - Denis R. Joanisse
- Département de kinésiologie, Faculté de médecine, Université Laval, Québec, Canada
- Centre de recherche de l’institut Universitaire de cardiologie et pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Canada
| | - Pascale Mauriège
- Département de kinésiologie, Faculté de médecine, Université Laval, Québec, Canada
- Centre de recherche de l’institut Universitaire de cardiologie et pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Canada
| |
Collapse
|
49
|
Zhang X, Liu Z, Li W, Kang Y, Xu Z, Li X, Gao Y, Qi Y. MAPKs/AP-1, not NF-κB, is responsible for MCP-1 production in TNF-α-activated adipocytes. Adipocyte 2022; 11:477-486. [PMID: 35941819 PMCID: PMC9367654 DOI: 10.1080/21623945.2022.2107786] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Obesity is associated with the infiltration of monocytes/macrophages into adipose
tissue in which MCP-1 plays a crucial role. But the regulatory mechanism of
MCP-1 expression in adipocytes is not well defined. Our results demonstrated that TNF-α induced abundant MCP-1 production in adipocytes, including 3T3-L1 pre- (≈ 9 to 18-fold), mature adipocytes (≈ 4 to 6-fold), and primary adipocytes(< 2-fold), among which 3T3-L1 pre-adipocytes showed the best reactiveness. Thus, 3T3-L1 pre-adipocytes were used for the most of following experiments. At the transcriptional level, TNF-α (20 ng/mL) also promoted the mRNA expression of MCP-1. It is well recognized that the engagement of TNF-α with its receptor can trigger both NF-κB and AP-1 signalling, which was also confirmed in our study (5-fold and 2-fold). Unexpectedly and counterintuitively, multiple NF-κB inhibitors with different mechanisms failed to suppress TNF-α-induced MCP-1 production, but rather the inhibitors for any one of MAPKs (JNK, ERK and p38) could do. This study, for the first time, reveals that MAPKs/AP-1 but not NF-κB signalling is responsible for MCP-1 production in TNF-α-activated adipocytes. These findings provide important insight into the role of AP-1 signalling in adipose tissue, and may lead to the development of therapeutical repositioning strategies in metaflammation. Abbreviations:
AP-1, activator protein-1; CHX, cycloheximide; IR, insulin resistance; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor κB; RT-qPCR, quantitative real-time PCR; T2DM, type 2 diabetes mellitus; TRE, triphorbol acetate-response element.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Zhuangzhuang Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Wenjing Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Yuan Kang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Zhenlu Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Ximeng Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Yuan Gao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Yun Qi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| |
Collapse
|
50
|
The Effect of Bariatric Surgery on Circulating Levels of Monocyte Chemoattractant Protein-1: A Systematic Review and Meta-Analysis. J Clin Med 2022; 11:jcm11237021. [PMID: 36498595 PMCID: PMC9738353 DOI: 10.3390/jcm11237021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Background: MCP-1 (monocyte chemoattractant protein) plays an important role in early phases of atherogenesis as well as in plaque destabilization, which causes cardiovascular events to play an important role in low-grade inflammation. Obesity, particularly extreme obesity, is a pivotal risk factor for atherosclerosis and many other diseases. In the early stages, bariatric surgery might stop or slow atherogenesis by suppressing inflammation, but also in later stages, preventing plaque destabilization. The aim of this meta-analysis was to provide an answer as to whether bariatric surgery has a significant effect on circulating MCP-1 level or not. Methods: A systematic literature search in PubMed, Scopus, Embase, and Web of Science was performed from inception to 1 January 2022. Meta-analysis was performed using Comprehensive Meta-Analysis (CMA) V2 software. In order to heterogeneity compensation of studies in terms of study design and treatment duration, the characteristics of the studied populations random-effects model and the generic inverse variance weighting method were used. To investigate the relationship with the estimated effect size, a random-effect meta-regression model was used. To assess the exitance of publication bias in the meta-analysis, the funnel plot, Begg's rank correlation, and Egger's weighted regression tests were used. Results: Meta-analysis of 25 studies with 927 subjects included demonstrated a significant decrease of MCP-1 concentration after bariatric surgery. The data of meta-regression did not indicate any association between the alterations in body mass index (BMI) and absolute difference in MCP-1 levels, but a linear relationship between the changes in MCP-1 and length of follow-up was proven. Conclusions: Bariatric surgery significantly decreases MCP-1 concentration, but there was no association between the changes in BMI and absolute difference in MCP-1 levels before and after the surgery.
Collapse
|